US20080274195A1 - Compositions and Methods for Making and Using Nanoemulsions - Google Patents

Compositions and Methods for Making and Using Nanoemulsions Download PDF

Info

Publication number
US20080274195A1
US20080274195A1 US11/996,016 US99601606A US2008274195A1 US 20080274195 A1 US20080274195 A1 US 20080274195A1 US 99601606 A US99601606 A US 99601606A US 2008274195 A1 US2008274195 A1 US 2008274195A1
Authority
US
United States
Prior art keywords
nanoemulsion
compound
particles
microfluidized
approximately
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/996,016
Inventor
Robert Nicolosi
Thomas Wilson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
UMass Lowell
Original Assignee
UMass Lowell
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by UMass Lowell filed Critical UMass Lowell
Priority to US11/996,016 priority Critical patent/US20080274195A1/en
Assigned to UNIVERSITY OF MASSACHUSETTS LOWELL reassignment UNIVERSITY OF MASSACHUSETTS LOWELL ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WILSON, THOMAS A, NICOLOSI, ROBERT
Publication of US20080274195A1 publication Critical patent/US20080274195A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/113Multiple emulsions, e.g. oil-in-water-in-oil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F23/00Mixing according to the phases to be mixed, e.g. dispersing or emulsifying
    • B01F23/40Mixing liquids with liquids; Emulsifying
    • B01F23/41Emulsifying
    • B01F23/413Homogenising a raw emulsion or making monodisperse or fine emulsions
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/773Nanoparticle, i.e. structure having three dimensions of 100 nm or less
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis
    • Y10S977/906Drug delivery

Definitions

  • the present invention relates to the field of nanoemulsions.
  • nanoemulsions are made using high shear stress technology.
  • the invention comprises uniform microfluidized nanoemulsions.
  • the uniform nanoemulsion comprises a compound such as a pharmaceutical, nutraceutical, or cosmeceutical.
  • the uniform nanoemulsion comprises improved pharmacokinetic parameters when compared to conventional nanoparticulate compositions and/or nanoemulsions.
  • the present invention contemplates a method of making a bacteria-resistant nanoemulsion.
  • Micro/nanoemulsion technology has substantial commercial value. In relation to the nutraceutical area alone, the market value is estimated as a 250 billion dollar business world-wide. Consequently, the ability to incorporate lipid soluble nutraceuticals into beverages (the fastest-growing component of the food industry) as well as low or no fat foods is of important interest.
  • nanoemulsions that: i) has improved temperature and pH stability; ii) improved bioavailability; and iii) improved shelf-life due to microbial resistance.
  • nanoemulsions should be relatively easy and inexpensive to prepare.
  • the present invention relates to the field of nanoemulsions.
  • the nanoemulsion is made using a high shear stress technology.
  • the invention comprises uniform microfluidized nanoemulsions.
  • the uniform nanoemulsion comprises a compound such as a pharmaceutical, nutraceutical, or cosmeceutical.
  • the uniform nanoemulsion comprises improved pharmacokinetic parameters when compared to conventional nanoparticulate compositions and/or nanoemulsions.
  • the present invention contemplates a method of making a bacteria-resistant nanoemulsion.
  • the present invention contemplates a nanoemulsion comprising a population of particles having maximum and minimum diameters, wherein the difference between said maximum and minimum diameters does not exceed 100 nm.
  • the present invention contemplates a nanoemulsion comprising a population of particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers.
  • the particles encapsulate a compound.
  • the compound is a pharmaceutical.
  • the compound is a nutraceutical.
  • the present invention contemplates a nanoemulsion comprising a first and second population of particles, wherein the majority of particles in said first population have diameters between approximately 10 and approximately 20 nanometers, wherein the majority of particles in said second population have diameters between approximately 40 and approximately 80 nanometers, wherein said nanoemulsion is uncontaminated by particles having diameters larger than 110 nanometers.
  • the particles encapsulate a compound.
  • the compound is a pharmaceutical.
  • the compound is a nutraceutical.
  • a nanoemulsion comprising a population of particles having diameters between approximately 50 and approximately 150 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 160 nanometers.
  • the particles encapsulate a compound.
  • the compound is a pharmaceutical.
  • the compound is a nutraceutical.
  • the present invention contemplates a method, comprising: a) providing; i) a premix comprising a compound and a liquid dispersion medium, wherein said compound has a solubility greater than 30 mg/ml in said medium; and ii) a microfluidizer capable of maintaining at least 25,000 PSI; b) using a single pass exposure of said premix to said microfluidizer to create a population of nanoemulsion particles having diameters ranging approximately between 10-110 nm.
  • the dispersion medium is selected from the group consisting of aqueous media and oil-based media.
  • the aqueous media is selected from the group consisting of water, ringers solution, dextrose, and short chain alcohols.
  • the oil-based media is selected from the group including, but not limited to, saturated and unsaturated oils from vegetable and marine sources, silicone oils, mineral oils, and plant-derived oils.
  • the compound is selected from the group including, but not limited to, a plant sterol, cod liver oil, tocopherol, lecithin, lutein, zeaxanthin, and soy protein.
  • the present invention contemplates a method, comprising: a) providing; i) a heated dispersion medium; ii) a compound having substantial solubility in said medium; and iii) a microfluidizer capable of making a uniform nanoemulsion from said medium; b) adding said compound to said medium at a temperature of at least 70° C. to create a premix; and c) microfluidizing said premix at a pressure of at least 25,000 PSI to create said nanoemulsion having particle diameters ranging between 10-110 nm.
  • said dispersion medium is selected from the group consisting of soybean oil and water.
  • said dispersion medium is heated to at least 65° C.
  • said compound may be selected from the group comprising a plant sterol, cod liver oil, tocopherol, lecithin, lutein, zeaxanthin, lycopene, whey protein, and soy protein.
  • the nanoemulsion encapsulates the compound.
  • 86% of said particle diameters have a 54 nm average diameter.
  • 14% of said particles diameters have a 16 nm average diameter.
  • 82% of said particle diameters have a 64 nm average diameter.
  • 17% of said particle diameters have a 19 nm average diameter.
  • 78% of said particle diameters have a 88 nm average diameter.
  • 22% of said particle diameters have a 27 nm average diameter. In one embodiment, 84% of said particle diameters have a 90 nm average diameter. In one embodiment, 16% of said particle diameters have a 23 nm average diameter. In one embodiment, 80% of said particle diameters have a 55 nm average diameter.
  • the present invention contemplates a method, comprising: a) providing; i) a premix comprising a compound, a first antioxidant, a second antioxidant, and an aqueous dispersion medium, wherein said compound has a solubility greater than 30 mg/ml in said medium; and iii) a microfluidizer capable of maintaining at least 25,000 PSI; c) using a single pass exposure of said premix to said microfluidizer to create a population of nanoemulsion particles having diameters ranging from between approximately 40-110 nm, wherein said particle diameter remains stable for at least four months.
  • the method further comprises pasteurizing said population of nanoemulsion particles wherein said particle diameters remain stable.
  • the method further comprises freezing said population of nanoemulsion particles wherein said particle diameters remain stable.
  • the present invention contemplates a method, comprising: a) providing; i) a stable aqueous dispersion medium comprising a first antioxidant; ii) a solution comprising natural emulsifiers; ii) a compound having substantial solubility in said medium comprising a second antioxidant; and iii) a microfluidizer capable of making a uniform nanoemulsion from said medium; b) adding said compound and said solution to said medium and heating to a temperature of at least 50° C.
  • the nanoemulsion encapsulates the compound.
  • the method further comprises pasteurizing said nanoemulsion wherein said particle diameters remain stable.
  • the method further comprises freezing said nanoemulsion wherein said particle diameters remain stable.
  • said solution comprises milk.
  • said compound comprises DHA fish oil.
  • said pasteurization comprises exposing said nanoemulsions to 75° C. for thirty seconds.
  • said freezing comprises exposing said nanoemulsions to ⁇ 4° C. for 24 hours.
  • the present invention contemplates a method, comprising; a) providing; i) a subject refractory to an administered compound at a therapeutically effective amount; ii) a nanoemulsion comprising a population of particles encapsulating said compound, wherein said particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers; b) delivering said nanoemulsion to said patients under conditions such that said compound bioavailability is improved and wherein said compound is therapeutically effective.
  • the improved bioavailability comprises pharmacokinetic parameters selected from the group consisting of decreased T max , increased C max , and increased AUC.
  • the delivering comprises a method selected from the group consisting of oral, transdermal, intravenous, intraperitoneal, intramuscular, and subcutaneous.
  • the nanoemulsion comprises a plant sterol. In one embodiment, the nanoemulsion comprises lycopene.
  • the present invention contemplates a method for improving a nanoemulsion bioavailability comprising providing a uniform microfluidized nanoemulsion and delivering the uniform nanoemulsion to a subject.
  • the subject comprises a mammal.
  • the nanoemulsion encapsulates a compound.
  • the nanoemulsion is delivered by oral administration.
  • the nanoemulsion is delivered by methods including, but not limited to, transdermally, intravenously, intraperitoneally, intramuscularly or subcutaneously.
  • said improved bioavailability comprises pharmacokinetic parameters selected from the group consisting of decreased T max , increased C max , and increased AUC.
  • said nanoemulsion is formulated for oral administration.
  • said nanoemulsion comprises a plant sterol.
  • said nanoemulsion comprises lycopene.
  • the present invention contemplates a nanoemulsion having bacteria-resistant properties, wherein said nanoemulsion comprises a population of particles encapsulating said compound, wherein said particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers.
  • the nanoemulsion resists bacterial growth for at least three months.
  • the bacterial-resistant properties comprise shear-force induced cell lysis.
  • the bacterial-resistant properties comprise an oxidizing environment.
  • the nanoemulsion is sterile.
  • the present invention contemplates a uniform microfluidized nanoemulsion comprising bacteria-resistant properties.
  • said nanoemulsion resists bacterial growth for at least three months.
  • the nanoemulsion comprises particles having a diameter distribution of between 10-110 nm.
  • said bacterial-resistant properties comprise shear-force induced cell lysis.
  • the nanoemulsion is sterile.
  • the present invention contemplates a method, comprising: a) providing; i) a premix comprising a compound and a liquid dispersion medium; and ii) a device capable of creating a continuous turbulent flow under high pressure; b) using said device to create a population of nanoemulsion particles having uniform diameter.
  • the dispersion medium is selected from the group consisting of aqueous media and oil-based media.
  • the aqueous media is selected from the group consisting of water, saline solution, ringers solution, dextrose, and short chain alcohols.
  • the oil-based media is selected from the group consisting of saturated and unsaturated oils from vegetable and marine sources, silicone oils, and mineral oils.
  • the compound is selected from the group consisting of a plant sterol, cod liver oil, tocopherol, lecithin, lutein, zeaxanthin, and soy protein.
  • microfluidized refers to an instrument or a process that utilizes a continuous turbulent flow at high pressure including, but not limited to, a microfluidizer or other like device that may be useful in creating a uniform nanoemulsion.
  • microfluidizing may create a uniform nanoemulsion comprising a pharmaceutical, nutraceutical, or cosmeceutical from a premix within a thirty (30) second time frame (typically referred to a single pass exposure).
  • a microfluidizer may be operated at a pressure of approximately 25,000 PSI to generate a uniform nanoemulsion.
  • uniform nanoemulsion refers to any emulsion comprising any specified range of particle diameter sizes wherein the difference between the minimum diameter and maximum diameters do not exceed approximately 600 nm, preferably approximately 300 nm, more preferably approximately 200 nm, but most preferably approximately 100 nm (i.e., for example, microfluidization, as contemplated herein, produces a uniform nanoemulsion having a range of approximately 10-110 nm and is referred to herein as a uniform microfluidized nanoemulsion).
  • the total particle distribution i.e., 100%
  • a particle diameter distribution where less than 3% is outside the specified range of particle diameter sizes is still contemplated herein as a uniform nanoemulsion.
  • population refers to any mixture of nanoemulsion particles having a distribution in diameter size.
  • a population of nanoemulsion particles may range is particle diameter from between approximately 10-10 nm.
  • nanoparticle refers to any particle having a diameter of less than 300 nanometers (nm), as defined by the National Science Foundation or preferably less than 100 nm, as defined by the National Institutes of Health. Most conventional techniques create nanoparticle compositions with an average particle diameter of approximately 300 nanometers (nm) or greater.
  • Oil-based liquids may include, but not limited to; saturated and unsaturated oils from vegetable and marine sources including, but not limited to, soybeans, safflowers, olives, corn, cottonseeds, linseed, safflower, palm, peanuts, flaxseeds, sunflowers, rice bran, sesame, rapeseed, cocoa butter etc., and mixtures thereof; silicone oils; and mineral oils.
  • aqueous media may include, but are not limited to, water, saline solutions, short chain alcohols, 5% dextrose, Ringer's solutions (lactated Ringer's injection, lactated Ringer's plus 5% dextrose injection, acylated Ringer's injection), Normosol-M, Isolyte E, and the like; and synthetic and/or natural detergents having high surfactant properties, deoxycholates, cyclodextrins, chaotropic salts and ion pairing agents etc., and mixtures thereof.
  • compound refers to any pharmaceutical, nutraceutical, or cosmeceutical (i.e., for example, organic chemicals, lipids, proteins, oils, vitamins, crystals, minerals etc.) that are substantially soluble in a dispersion medium.
  • pharmaceutical, nutraceutical, or cosmeceutical i.e., for example, organic chemicals, lipids, proteins, oils, vitamins, crystals, minerals etc.
  • substantially soluble refers to any compound that dissolves into a dispersion medium to a concentration greater than 30 mg/ml.
  • the dispersion medium is heated while the compound is being dissolved.
  • premixe refers to any mixture that is subsequently used to generate a nanoparticulate composition or a uniform microfluidized nanoemulsion.
  • premixes typically contain a liquid dispersion medium and a compound, and optionally, an emulsifier and/or an antioxidant.
  • stable refers to any population of nanoemulsion particles whose diameters stay within the range of approximately 10-110 nm over a prolonged period of time (i.e., for example, one (1) day to twenty-four (24) months, preferably, two (2) weeks to twelve (12) months, but more preferably two (2) months to five (5) months). For example, if a population of nanoemulsion particles is subjected to prolonged storage, temperature changes, and/or pH changes whose diameters stay within a range of between approximately 10-110 nm, the nanoemulsion is stable.
  • bacteria-resistant refers to the lack of observable bacterial growth.
  • sterile refers to a nanoemulsion that contains no living bacterial cells.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts refers to derivatives wherein the parent compound is modified by making acid or base salts thereof.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, and the like
  • organic acids such as acetic, propionic, succinic, glycolic, stearic,
  • terapéuticaally effective amount as used herein, with respect to a drug dosage, shall mean that dosage that provides the specific pharmacological response for which the drug is administered or delivered to a significant number of subjects in need of such treatment. It is emphasized that ‘therapeutically effective amount,’ administered to a particular subject in a particular instance will not always be effective in treating the diseases described herein, even though such dosage is deemed a “therapeutically effective amount” by those skilled in the art. Specific subjects may, in fact, be “refractory” to a “therapeutically effective amount”. For example, a refractory subject may have a low bioavailability such that clinical efficacy is not obtainable. It is to be further understood that drug dosages are, in particular instances, measured as oral dosages, or with reference to drug levels as measured in blood.
  • refractory refers to any subject that does not respond with an expected clinical efficacy following the delivery of a compound as normally observed by practicing medical personnel.
  • delivering refers to any route for providing a pharmaceutical or a nutraceutical to a subject as accepted as standard by the medical community.
  • routes of delivering or administering that include, but are not limited to, oral, transdermal, intravenous, intraperitoneal, intramuscular, or subcutaneous.
  • subject refers to any animal to which an embodiment of the present invention may be delivered or administered.
  • a subject may be a human, dog, cat, cow, pig, horse, mouse, rat, gerbil, hamster etc.
  • encapsulate refers to any compound that is completely surrounded by a protective material.
  • a compound may become encapsulated by a population of nanoemulsion particle formation during microfluidization.
  • a dietary source i.e., for example, a fortified food or a dietary supplement
  • a dietary source i.e., for example, a fortified food or a dietary supplement
  • compositions including, but not limited to, a cream, oil, foam, spray, liquid etc.
  • Cosmeceuticals may include categories such as, but not limited to, carotenoids, phenolic compounds, or water soluble antioxidants.
  • FIG. 1 presents exemplary data showing the particle diameter distribution of a microfluidized plant sterol nanoemulsion population three (3) months after preparation.
  • FIG. 1A presents exemplary data showing the particle diameter distribution of a microfluidized plant sterol nanoemulsion three (3) months after preparation.
  • FIG. 2 presents exemplary data showing the particle diameter distribution of a microfluidized cod liver oil nanoemulsion population four (4) months after preparation.
  • FIG. 3 presents exemplary data showing the particle diameter distribution of a microfluidized tocopherol nanoemulsion population five (5) months after preparation.
  • FIG. 4 presents exemplary data showing the particle diameter distribution of a microfluidized lutein/zeaxanthin nanoemulsion population.
  • FIG. 5 presents exemplary data showing the particle diameter distribution of a microfluidized soy protein nanoemulsion population.
  • FIG. 6 presents exemplary data showing the particle diameter distribution of a microfluidized whey protein nanoemulsion population.
  • FIG. 7 presents exemplary data showing the particle diameter distribution of a microfluidized orange juice/plant sterol/lutein nanoemulsion population.
  • FIG. 8 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/water nanoemulsion population two (2) months after preparation.
  • FIG. 9 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/milk nanoemulsion population three (3) weeks after preparation.
  • FIG. 10 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/milk/tocopherol nanoemulsion population.
  • FIG. 11 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/milk/tocopherol nanoemulsion population after pasteurization.
  • FIG. 12 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/milk/tocopherol nanoemulsion population after a freeze-thaw process.
  • FIG. 13 presents exemplary data of gerbil plasma lycopene levels when fed a lycopene-enriched diet.
  • FIG. 14 presents exemplary data of gerbil plasma lycopene levels when fed a microfluidized lycopene nanoemulsion diet.
  • FIG. 15 presents one embodiment of an anti-bacterial property generated during the preparation of a microfluidized plant sterol nanoemulsion.
  • FIG. 16 presents exemplary data showing the particle diameter distribution of a microfluidized plant sterol nanoemulsion population used in Example 12.
  • FIG. 17 presents exemplary data showing that a microfluidized plant sterol nanoemulsion diet is more effective in reducing plasma LDL-C in hypercholesterolemic hamsters than either a micronized plant sterol diet or a crystalline plant sterol diet for four (4) weeks.
  • FIG. 18 presents exemplary data comparing premix cholesterol particle diameter distributions from: Panel A: Tween® 80/Water as per the '118 patent; and Panel B: Oil/Lecithin/Tween® 80/Water as contemplated by one embodiment of the present invention.
  • FIG. 19 presents exemplary data comparing microfluidized cholesterol nanoemulsion particle diameter distributions from: Panel A: Tween® 80/Water as per the '118 patent using repeated microfluidization passes; and Panel B: Oil/Lecithin/Tween® 80/Water as contemplated by one embodiment of the present invention using a single microfluidization pass.
  • FIG. 20 presents exemplary data comparing microfluidized cholesterol nanoemulsion particle diameter distributions from: Panel A: Tween® 80/Water as per the '118 patent using a single pass exposure; and Panel B: Oil/Lecithin/Tween® 80/Water as contemplated by one embodiment of the present invention using a single pass exposure.
  • the present invention relates to the field of nanoemulsions.
  • the nanoemulsion is created by a high shear stress technology.
  • the invention comprises uniform microfluidized nanoemulsions.
  • the uniform nanoemulsion comprises a compound such as a pharmaceutical, nutraceutical, or cosmeceutical.
  • the uniform nanoemulsion comprises improved pharmacokinetic parameters when compared to conventional nanoparticulate compositions and/or nanoemulsions.
  • the present invention contemplates a method of making a bacteria-resistant nanoemulsion.
  • Nanoemulsion nutraceutical delivery is generally directed to pharmaceuticals.
  • Nanoemulsion nutraceutical delivery has received little attention.
  • one nanoemulsion system contains plant sterols.
  • Bruce et al. “Method for producing dispersible sterol and stanol compounds” U.S. Pat. No. 6,387,411 (2002) (herein incorporated by reference).
  • This technology uses a grinding method to produce the nanoemulsions, and consequently, the particle diameter is at least six (6) times greater than contemplated herein.
  • this diameter difference offers particular advantages in stability and efficacy (infra).
  • the '411 patent does not disclose the incorporation of absorbable micronutrients.
  • Cosmeceuticals may comprise, for example; carotenoids including, but not limited to, ⁇ -carotene, ⁇ -carotene, ⁇ -cryptoxanthin, lycopene, crocetin, fucoxanthin, halocynthiaxanthin, canthaxanthin, astraxanthin, lutein, or zeaxanthin; phenolic compounds including, but not limited to, quercetin, rutin, myricetin, kaemferol, catechin, epigallocatechin, epicatechin, reservatrol, tocopherol, ferulate, ubiquinol-10, soy isoflavones such as genestein, daidzein, alpha lipoic acid, anthocyanins, ellagic tannins, gallic or ellagic acids; or water soluble anti-oxidants such as ascorbic acid,
  • the present invention is directed to populations of nanoparticles or nanoemulsions comprising an oral delivery vehicle for all absorbable (i.e., for example, fat-soluble) nutrients including, but not limited to, fatty acids, carotenoids, tocopherols, tocotrienols, and coenzyme-Q. Delivery methods, however, are not limited to oral and include, but are not limited to, transdermal, intravenous, intraperitoneal, intramuscular, or subcutaneous.
  • the carotenoids include, but are not limited to, lutein and zeaxanthin.
  • the present invention is also directed to populations of nanoparticles or nanoemulsions comprising an oral delivery vehicle for all non-absorbable (i.e., for example, fat soluble) plant sterol compounds including, but not limited to, phytosterols and phytostanols.
  • the compounds are encapsulated by the nanoparticles or nanoemulsions.
  • common emulsifying agents are used to prepare the nanoemulsions.
  • the emulsifying agents include, but are not limited to, phospholipids, fatty acid monoglycerides, fatty acid diglycerides, or polysorbates.
  • the present invention also contemplates that certain nanoemulsion embodiments of the present invention comprise a surface-to-volume ratio that results in an improved bioavailability over current methods and compositions known in the art.
  • the present invention also contemplates that certain nanoemulsion embodiments of the present invention are resistant to microbiological growth. Although it is not necessary to understand the mechanism of an invention, it is believed that the microfluidization process comprises a high sheer stress and/or creates an oxidizing environment, thereby disrupting microbial integrity and/or preventing microbial growth.
  • Nanoemulsions have been generated by a variety of methods. In particular, these methods provide a wide variation in particle diameter and require organic solvents and or polymers. When these known nanoemulsions are considered for an oral drug or nutrient delivery system, issues of biocompatibility and physiological side effects become an important issue.
  • the present invention contemplates a method of making a nanoemulsion comprising a continuous turbulent flow at high pressure.
  • the high pressure turbulent flow comprises microfluidization.
  • a uniform nanoemulsion is generated from a premix using a single pass exposure (i.e., for example, within a thirty (30) second time frame).
  • the uniform nanoemulsion comprises a population of particles whose difference between the minimum and maximum diameters does not exceed approximately 100 nm.
  • a uniform nanoemulsion is generated using a pressure of at least 25,000 PSI.
  • the present invention contemplates a method of making uniform microfluidized nanoemulsions without organic solvents or polymers.
  • the microfluidized nanoemulsion is made from a suspension.
  • the microfluidized nanoemulsion is made from a microemulsion.
  • the present invention contemplates a uniform microfluidized nanoemulsion using compounds that are substantially soluble in a liquid dispersion medium.
  • the nanoemulsion encapsulated the compounds.
  • the compounds comprise pharmaceuticals and/or nutraceuticals.
  • nutraceuticals and dietary supplements are disclosed, for example, in Roberts et al., Nutriceuticals: The Complete Encyclopedia of Supplements, Herbs, Vitamins, and Healing Foods (American Nutriceutical Association, 2001), which is specifically incorporated by reference. Dietary supplements and nutraceuticals are also disclosed in Physicians' Desk Reference for Nutritional Supplements, 1st Ed. (2001) and The Physicians' Desk Reference for Herbal Medicines, 1st Ed. (2001), both of which are also incorporated by reference.
  • a nutraceutical or dietary supplement, also known as a phytochemical or functional food is generally any one of a class of dietary supplements, vitamins, minerals, herbs, or healing foods that have medical or biological 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, amino acids (e.g., glutamine, arginine, iso-leucine, leucine, lysine, methionine, phenylalanine, 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.”
  • these compounds include, but are not limited to, naturally occurring oils, fatty acids, and proteins.
  • a naturally occurring oil comprises fish oil (i.e., for example, cod liver oil).
  • a naturally occurring fatty acid comprises an omega-3 (i.e., for example, DHA).
  • the nanoemulsion comprises little or no fat.
  • a naturally occurring protein comprises soy or whey.
  • the present invention contemplates a method of making a uniform microfluidized nanoemulsion comprising a population of particles whose diameter ranges from between 10-110 nm.
  • Microfluidization is a unique process that powers a single acting intensifier pump.
  • the intensifier pump amplifies the hydraulic pressure to the selected level which, in turn, imparts that pressure to the product stream.
  • Within the interaction chamber are specially designed fixed-geometry microchannels through which the product stream will accelerate to high velocities, creating high shear and impact forces that generates a uniform nanoemulsion as the high velocity product stream impinges on itself and on wear-resistant surfaces.
  • the intensifier pump completes its pressure stroke, it reverses direction and draws in a new volume of product. At the end of the intake stroke, it again reverses direction and drives the product at constant pressures, thereby repeating the process.
  • the product Upon exiting the interaction chamber, the product flows through an onboard heat exchanger which regulates the product to a desired temperature. At this point, the product may be recirculated through the system for further processing or directed externally to the next step in the process.
  • Cook et al. “Apparatus For Forming Emulsions” U.S. Pat. No. 4,533,254 (1985); and Cook et al., “Method Of Forming A Microemulsion” U.S. Pat. No. 4,908,154 (1990) (both herein incorporated by reference).
  • this previous process requires long-processing times (i.e., repeated microemulsifying cycles) thereby promoting heat build-up in the microfluidizer. Consequently, this early technique requires processing temperatures of less than 40° C.
  • This technique resulted in average nanoemulsion particle diameters of approximately 300 nm.
  • lower particle diameters i.e., less than 100 nm
  • Exemplary data presented herein has used the Bosch et al. process to produce a complete particle diameter distribution profile. See Example 13. These data show that the Bosch et al. technology cannot produce a uniform nanoemulsion as contemplated by the present invention.
  • nanoparticle composition as “particles consisting of a poorly soluble therapeutic or diagnostic agent having adsorbed onto, or associated with, the surface thereof a non-crosslinked surface stabilizer”.
  • Cooper et al. “Nanoparticulate Sterol Formulations And Novel Sterol Combinations” United States Patent Application Publication No. 2004/0033202 A1 (2004) (see pg 1 para 3) (herein incorporated by reference).
  • Cooper et al. discloses preparing nanoparticulate compositions using compounds that are poorly soluble in a liquid dispersion medium (i.e., water, oils, alcohols, glycols, etc.).
  • Cooper et al. defines a “poorly water-soluble” drug as having a solubility of less than about 30 mg/ml.
  • plant sterol nanoparticulate compositions comprising one or more sterols or stanols (i.e., sitosterol or phytosterols) are suggested in the art as having a particle diameter of less than 50 nm.
  • Cooper et al. does not use a microfluidizer nor present any data showing a capability of providing a uniform particle diameter ranging between 10-110 nm. Instead, Cooper et al. relies upon a more traditional milling process that does not produce a uniform particle diameter distribution ranging between 10-110 nm.
  • DYNO®-MILL KDL milling grinder
  • Glen Mills, Inc. Clifton, N.J.
  • the DYNO®-MILL is a versatile horizontal bead mill having applications ranging from paints and coatings to drug manufacturing and cell disruption for extracting proteins. Grinding to a mean diameter of 320 nm has been reported in research papers. Operation of the DYNO®-MILL is always wet, that is, the material to be ground is held in suspension in any suitable liquid.
  • a jacketed grinding chamber contains a series of agitators that are equally spaced along the length of a central shaft.
  • the jacket on the grinding chamber is used to control the temperature of the material being processed.
  • the chamber is secured at one end and cantilevers out over the shaft.
  • the bearing end contains a separator gap which has clearance tolerances that can be set as tight as 20 microns.
  • the chamber is filled to about 80% of its capacity with beads (i.e., PolyMill® 500; 500 ⁇ m diameter grinding beads). Depending on the specific application beads made from glass, ceramic, metals, tungsten carbide and other materials are available.
  • the process material is now introduced into the chamber. When the chamber is full of material and beads, the machine is switched on and the agitator discs rotate forcing the beads to impact over and over with the process material with hurricane-like force. This action of having thousands of separate impacts produces rapid and consistent size reduction. Batch and continuous processing can be handled in the same mill by changing the grinding chamber and the gap setting.
  • Cooper et al. is limited to a plant sterol nanoparticulate composition where 90% of the particle diameters are below 187 nm. The actual particle diameter distribution, however, is not presented. In one embodiment, the present invention contemplates that the technology described by Cooper et al. cannot produce a uniform particle diameter distribution ranging between 10-110 nm. See Example 14. Unlike some embodiments of the present invention, Cooper et al. has not considered methods to make a nanoparticulate composition that include a heating process. In fact, Cooper et al. presents a discussion concluding that preparing a plant sterol nanoparticulate composition using a process that includes heating is not desirable and problematic. Some embodiments of the present invention have solved those problems.
  • emulsification a physical process termed “emulsification”.
  • An emulsion is traditionally defined in the art “as a system . . . consisting of a liquid dispersed with or without an emulsifier in an immiscible liquid usually in droplets of larger than colloidal size” Medline Plus Online Medical Dictionary, Merriam Webster (2005). Consequently, as the art developed emulsifiers capable of generating smaller and smaller diameter particles, the terms “microemulsion” and “nanoemulsion” became known.
  • a microemulsion is one thousand-fold greater in diameter than a nanoemulsion.
  • particle diameter distributions may vary widely in a non-controlled emulsification process creating considerable overlap between the nanoemulsion and microemulsion technologies.
  • the present invention contemplates a premix comprising a compound substantially soluble (i.e., for example, greater than 30 mg/ml) in a liquid dispersion medium (i.e., for example, a heated liquid dispersion medium) and, optionally, common emulsifying agents including, but not limited to, phospholipids, fatty acid monoglycerides, fatty acid diglycerides, or polysorbates.
  • a nanoemulsion is created by exposing a premix to a continuous turbulent flow at a high pressure, wherein the pressure is at least 25,000 PSI.
  • the high pressure turbulent flow comprises microfluidization.
  • the nanoemulsion comprises particles encapsulating pharmaceuticals or nutraceuticals. In one embodiment, the nanoemulsion comprises a uniform nanoemulsion having stable particles. In one embodiment, the microfluidization comprises a single pass exposure (i.e., for example, approximately thirty (30) seconds). In one embodiment, a uniform plant sterol microfluidized nanoemulsion has an improved low density lipoprotein cholesterol lowering efficacy.
  • Oral drug administration is a common method for providing pharmaceuticals and nutraceuticals to any subject.
  • the contemplated methods of delivering a nanoemulsion is not limited to oral and include, for example, transdermal, intravenous, intraperitoneal, intramuscular, or subcutaneous routes of administration.
  • Oral administration is favored because the formulations (i.e., liquids or suspensions) are relatively inexpensive to produce and are well tolerated. Subsequent gastrointestinal absorption of the formulation's ingredients, however, is not as predictable.
  • the formulations must be compatible with the digestive system. Consequently, lipid-based drug delivery systems are known to be useful as carriers for many drug delivery systems.
  • lipid composition i.e., for example, molecular size and charge
  • pharmaceutical, nutraceutical, or cosmeceutical chemical structure i.e., molecular size and pH ionization
  • iii the overall health of the subject.
  • Lipids are generally categorized as physiologically non-absorbable or absorbable. It should be recognized that gastrointestinal absorption processes are unrelated to a compound's solubility properties.
  • the present invention contemplates compositions and methods related to uniform microfluidized nanoemulsions comprising either absorbable or non-absorbable lipids thereby improving their bioavailability.
  • Plant sterols, stanols, and triterpene alcohols are either not absorbed, or poorly absorbed, into the bloodstream following oral administration.
  • the present invention contemplates a method of making a uniform nanoemulsion (i.e., for example, microfluidized) comprising a non-absorbable lipid having substantial solubility in a liquid dispersion medium and, optionally, common emulsifying agents, such as phospholipids, fatty acid monoglycerides, fatty acid diglycerides, or polysorbates to formulate improved nanoemulsions.
  • the nanoemulsion comprises particle diameters ranging between 10-110 nm, thereby improving oral administration.
  • plant sterols such as ⁇ -sitosterol
  • ⁇ -sitosterol plant sterols
  • the presence of unabsorbed plant sterols in the gastrointestinal system inhibits the normal metabolism of cholesterol and, concomitantly, decreases blood cholesterol levels.
  • administration of twenty (20) gms of crystalline plant sterols can reduce plasma cholesterol levels approximately 10%.
  • non-absorbable lipids are advantageous as a nutraceutical because of a lack of side effects.
  • Side effects are routinely observed when using traditional pharmaceutical systemic cholesterol-lowering interventions (i.e., for example, HMG CoA reductase inhibitors or niacin).
  • HMG CoA reductase inhibitors or niacin i.e., HMG CoA reductase inhibitors or niacin.
  • plant sterols can be prescribed for the general population, including children for whom systemic interventions are rarely recommended. It is known that the consumption of adequate amounts of plant sterols will lower blood cholesterol levels.
  • the present invention contemplates improvements in currently known methods to deliver plant sterols or stanols.
  • the first known method involves dissolving the plant sterol in a vegetable oil-containing margarine to an efficacious level of plant sterol.
  • a free stanol or a sterol is increased by: i) interesterified with a fatty acid such oleate or linoleate; ii) mixed in vegetable oil; or iii) hydrogenated to produce margarine, plasma cholesterol can be reduced by approximately 30%.
  • this process can result in the consumption of up to approximately eighteen (18) grams of fat.
  • the present invention contemplates a method of making a beverage nanoemulsion that comprises plant sterols.
  • the method to make the beverage nanoemulsion may comprise a continuous turbulent flow at a high pressure.
  • the continuous turbulent high pressure flow comprises microfluidization.
  • the nanoemulsion beverage comprises an orange juice product.
  • the second known method comprises oral delivery of water-dispersible plant sterols (i.e., for example, a stanol not dissolved in fat) by incorporation micron-sized micelles (i.e., microemulsions having diameters of several thousand nanometers) which can be subsequently added to beverages or foods.
  • micron-sized micelles i.e., microemulsions having diameters of several thousand nanometers
  • Ostlund, Jr. “Sitostanol formulation to reduce cholesterol absorption and method for preparing and use of same” U.S. Pat. No. 5,932,562 (1999)(herein incorporated by reference).
  • cholesterol absorption was reduced by approximately 37%.
  • the present invention contemplates a nanoemulsion technology comprising a specific formulation and a microfluidization process that provides particle diameters from between 10-110 nm.
  • the nanoparticle has improved pH and temperature stability properties, thereby stabilizing the particle's integrity throughout the gastrointestinal system.
  • the third known method involves the oral delivery of plant sterols by producing a water dispersible sterol product.
  • These water dispersible products usually include emulsifying agents including, but not limited to, monoglycerides and polysorbates.
  • the present invention contemplates a microfluidizing nanoemulsion technology (i.e., for example, that produced by a continuous flow high pressure process) that improves the emulsification of these water-dispersible plant sterols into nanoemulsions having a particle diameter of approximately 40-60 nm.
  • any incorporated pharmaceutical, nutraceutical, or cosmeceutical has improved efficacy (i.e., for example, plasma cholesterol lowering by a plant sterol).
  • a smaller-sized plant sterol-containing nanoparticles contemplated by one embodiment of the present invention when compared to known micron-sized micelles or microemulsions, has an improved disruption of the normal micellar delivery of dietary cholesterol to the digestive tract.
  • This invention also relates to the use of nanoemulsions as an oral delivery vehicle for absorbable lipids including, but not limited to, fatty acids, carotenoids, tocopherols and other fat soluble vitamins, tocotrienols, and Coenzyme-Q.
  • the present invention contemplates a method to make a uniform microfluidized nanoemulsion comprising an absorbable lipid having substantial solubility in a liquid dispersion medium and, optionally, common emulsifying agents, such as phospholipids, fatty acid monoglycerides, fatty acid diglycerides, or polysorbates to formulate improved nanoemulsions.
  • the method comprises a step exposing a premix to a continuous turbulent flow at high pressure.
  • the pressure is at least 25,000 PSI.
  • the nanoemulsion comprises carotenoids, including, but not limited to, lutein and zeaxanthin.
  • the nanoemulsion comprises nanoparticles having a particle diameter ranging from 10-110 nm, thereby improving bioavailability.
  • nanoemulsion bioavailability is improved following oral, transdermal, intravenous, intraperitoneal, intramuscular or subcutaneous delivery.
  • the present invention contemplates a method to treat or prevent macular degeneration (i.e., a major cause of blindness in people of 65) providing an improved nanoemulsion comprising at least one carotenoid.
  • the carotenoid is selected from the group comprising lutein or zeaxanthin.
  • lipid bioavailability i.e., for example, the percentage of the dose absorbed.
  • Factors influencing bioavailability may include, but are not limited to, food processing methods, food matrix, and physiological solubility in naturally-occurring micelles (i.e., for example, the lipid micellular transport system).
  • Fat-soluble nutrients can be incorporated into high fat-containing vegetable oils for dispersal into a fat matrix (i.e., for example a micron-sized micelle).
  • the micelle solubilizes the lipid-soluble nutrient thereby allowing absorption by the small intestine.
  • cholesterol absorption inhibition is increased up to three-fold.
  • Ostlund et al. “Sitostanol administered in lecithin micelles potently reduces cholesterol absorption in humans” Am J Clin Nutr 70:826-831 (1999).
  • the present invention contemplates a nanoemulsion produced by a continuous turbulent flow at high pressure having improved pharmacokinetic properties when compared to conventional nanoparticulate compositions and/or nanoemulsions currently known in the art. It is known that nanoparticles deliver and/or release drugs (i.e., for example, norflaxin) and/or proteins (i.e., for example, serum albumin) more effectively than microparticles.
  • drugs i.e., for example, norflaxin
  • proteins i.e., for example, serum albumin
  • One embodiment of the present invention contemplates a uniform microfluidized nanoemulsion having improved pharmacokinetic properties when compared to conventional nanoparticulate compositions and/or nanoemulsions currently known in the art.
  • One advantage of uniform microfluidized nanoemulsions comprises a narrow particle diameter range (i.e., for example, 10-110 nm).
  • Most conventional nanoparticle compositions and/or nanoemulsions currently known have a wide distribution of particle diameters that interfere with the improved efficacies and bioavailabilities of the smaller sized particles.
  • the present invention has solved the problem of generating nanoemulsions with highly variable particle diameters and provides a more uniformly small-sized nanoemulsions (i.e., for example, a uniform nanoemulsion comprising stable particles). Consequently, these uniform nanoemulsions provide improved pharmacokinetic parameters when compared to conventional nanoparticle compositions and/or nanoemulsions currently known in the art independent of the mode of delivery which includes, but is not limited to, oral, transdermal, intravenous, intraperitoneal, intramuscular, subcutaneous, etc.
  • nanoparticulate compositions or nanoemulsions are not ideal due to delayed onset of action.
  • a uniform microfluidized nanoemulsion as contemplated by the present invention exhibits faster therapeutic effects.
  • peak plasma concentrations of an incorporated compound can be obtained in less than about 2 hours, preferably less than about 1 hour, more preferably less than about 30 minutes, but most preferably between 1 and 15 minutes.
  • the recommended total daily dose of most pharmaceuticals and nutraceuticals are administered in divided doses. It is known in the art that a single daily dose may be preferable to multiple dose each day. For example, in studies of adults with partial onset seizures, a daily dose of 200 mg/day has inconsistent effects and is less effective than 400 mg/day. See Physicians' Desk Reference, 57.sup.th Edition, pp. 2502 (2003).
  • some uniform microfluidized nanoemulsions of the present invention may be administered less frequently, at lower doses, and in dosage forms such as liquid dispersions, powders, sprays, solid re-dispersible dosage forms, ointments, creams, etc.
  • dosage forms such as liquid dispersions, powders, sprays, solid re-dispersible dosage forms, ointments, creams, etc.
  • Exemplary types of formulations useful in the present invention include, but are not limited to, liquid dispersions, gels, aerosols (pulmonary and nasal), ointments, creams, solid dose forms, etc. of any pharmaceutical, nutraceutical, or cosmeceutical.
  • Lower dosages can be used because the smaller particle diameters of embodiments of the present invention ensure more complete absorption.
  • the present invention contemplates a therapeutically effective amount of a uniform microfluidized nanoemulsion having 1 ⁇ 6, 1 ⁇ 5, 1 ⁇ 4, 1 ⁇ 3, or 1 ⁇ 2 of the therapeutically effective amount of a conventional pharmaceutical, nutraceutical, or cosmeceutical formulations.
  • a liquid dosage form of a conventional nanoparticulate or nanoemulsion composition would be expected to be a relatively large volume, highly viscous substance which would not be well accepted by subject populations. Moreover, viscous solutions can be problematic in parenteral administration because these solutions require a slow syringe push and can stick to tubing. In addition, conventional formulations of poorly water-soluble active agents tend to be unsafe for intravenous administration techniques, which are used primarily in conjunction with highly water-soluble substances. Embodiment contemplated by the present invention solves this problem by utilizing a liquid dispersion medium in which the pharmaceutical, nutraceutical, or cosmeceutical is substantially soluble.
  • Liquid dosage forms of embodiments of a uniform microfluidized nanoemulsion provide significant advantages over a liquid dosage form of a conventional nanoparticulate or nanoemulsion.
  • the uniform microfluidized nanoemulsion comprises a low viscosity.
  • the uniform nanoemulsion comprises a silky texture.
  • Liquid formulations of uniform nanoemulsions contemplated by the present invention are easier to consume which is especially important when considering juvenile subjects, terminally ill subjects, and elderly subjects. Viscous or gritty formulations, and those that require a relatively large dosage volume, are not well tolerated by these subject populations.
  • Liquid oral dosage forms can be particularly preferably for subject populations who have difficulty consuming tablets, such as infants and the elderly.
  • the viscosities of liquid dosage forms of nanoparticulate topiramate according to the invention are preferably less than about 1/200, less than about 1/175, less than about 1/150, less than about 1/125, less than about 1/100, less than about 1/75, less than about fraction 1/50, or less than about 1/25 of a liquid oral dosage form of a conventional nanoparticulate composition or nanoemulsion at about the same concentration per ml.
  • the present invention contemplates a uniform microfluidized nanoemulsion that is not turbid.
  • turbid refers to the property of particulate matter that can be seen with the naked eye or that which can be felt as “gritty” when consumed.
  • Embodiments of nanoemulsions contemplated by the present invention can be poured out of or extracted from a container as easily as water, whereas a liquid dosage form of a conventional nanoparticulate or nanoemulsion composition is expected to exhibit notably more “sluggish” characteristics.
  • the present invention contemplates a uniform microfluidized nanoemulsion having an increased bioavailability and a smaller dose requirement as compared to prior conventional nanoparticulate compositions and nanoemulsions administered at the same dose.
  • Any pharmaceutical, nutraceutical, or cosmeceutical can have adverse side effects if administered at a specific dose for a specific duration.
  • lower doses which can achieve the same or better therapeutic effects as those observed with larger doses are desired.
  • Such lower doses may be realized with a uniform microfluidized nanoemulsion contemplated by the present invention due to greater bioavailability as compared to conventional nanoparticulate compositions and nanoemulsions; consequently smaller dose of pharmaceuticals and nutraceutical are likely required to obtain the desired therapeutic effect.
  • the relative bioavailability of pharmaceutical, nutraceutical, or cosmeceutical incorporated into a conventional nanoparticulate or nanoemulsion may be about 85% (i.e., as compared to a pure solution).
  • a uniform microfluidized nanoemulsion formulated into an oral pharmaceutical, nutraceutical, or cosmeceutical dosage form has a relative bioavailability preferably greater than about 85%.
  • the relative bioavailability is greater than about 90%, or greater than about 95%, or greater than about 98%.
  • the present invention also provides embodiments of uniform microfluidized nanoemulsions having incorporated pharmaceuticals and/or nutraceuticals having improved pharmacokinetic profiles when administered to mammalian subject.
  • the improved profile is compared to conventional nanoparticulate compositions and nanoemulsions.
  • an improved pharmacokinetic (pK) profile can have several different types of attributes.
  • an improved pK profile of a uniform microfluidized nanoemulsion may produce the same pK profile as a conventional nanoparticulate composition or nanoemulsion, but at a lower dose.
  • an improved pK profile requires less frequent dosing as compared to a conventional nanoparticulate composition or nanoemulsion.
  • an improved pK profile shows a faster onset of activity and/or greater quantity of drug absorbed (i.e., greater bioavailability) than conventional nanoparticulate compositions and nanoemulsions.
  • an improved pK profile allows a more effective and/or faster titration of the subject to therapeutic plasma levels.
  • the present invention contemplates certain embodiments of uniform microfluidized nanoemulsions comprising an improved pharmacokinetic profile as reflected by time-to-maximum-concentration (T max ), maximum-concentration (C max ), and/or area-under-curve (AUC) profiles.
  • T max time-to-maximum-concentration
  • C max maximum-concentration
  • AUC area-under-curve
  • an administered dose of a pharmaceutical, nutraceutical, or cosmeceutical incorporated into a uniform microfluidized nanoemulsion comprises a T max less than that of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • the T max is less than about 99%, less than about 90%, less than about 80%, less than about 70%, less than about 60%, less than about 50%, less than about 40%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, or less than about 10% of the T max of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • an administered dose of a pharmaceutical, nutraceutical, or cosmeceutical incorporated into a uniform microfluidized nanoemulsion comprises a C max greater than that of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • the C max is greater than about 5%, greater than about 10%, greater than about 15%, greater than about 20%, greater than about 30%, greater than about 40%, greater than about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, greater than about 100%, greater than about 110%, greater than about 120%, greater than about 130%, greater than about 140%, or greater than about 150% than the C max of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • an administered dose of a pharmaceutical, nutraceutical, or cosmeceutical incorporated into a uniform microfluidized nanoemulsion comprises an AUC greater than that of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • the AUC is greater than about 5%, greater than about 10%, greater than about 15%, greater than about 20%, greater than about 30%, greater than about 40%, greater than about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, greater than about 100%, greater than about 110%, greater than about 120%, greater than about 130%, greater than about 140%, or greater than about 150% than the AUC of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • the present invention contemplates a method of making a nanoemulsion having anti-bacterial properties.
  • the method comprises a step exposing a premix to a continuous turbulent flow at high pressure.
  • the anti-bacterial nanoemulsion is prepared by microfluidization.
  • the exposing comprises approximately thirty (30) seconds.
  • the exposing comprises a pressure of at least 25,000 PSI.
  • the anti-bacterial nanoemulsion comprises soy protein.
  • a powdered soy protein preparation was added to water thus creating a suspension. Then, a first aliquot of the suspension was added to a first container (i.e., for example, a cell culture falcon flask) that served as a control. A second aliquot of the suspension was microfluidized (supra) to create a nanoemulsion. The preparation was microfluidized in accordance with Example 5. The microfluidized nanoemulsion was then added to a second container. Both containers were refrigerated immediately and observed over the next several days. The control suspensions agglomerated and grew bacteria. See FIGS. 15A and 15B . In contrast, the microfluidized nanoemulsion containing the soy protein did not agglomerate or grow bacteria. See FIGS. 15C and 15D .
  • microfluidization sterilized the bacteria. It is further believed that the microfluidization shear stress resulted in a bacterial cell lysis thereby preventing further bacterial growth. Consequently, it is believed that microfluidization, as contemplated herein, produces a microbiologically sterile composition.
  • the present invention contemplates a nanoemulsion comprising an oxidizing environment produced by a method comprising a continuous turbulent flow at a high pressure.
  • the nanoemulsion comprises a uniform microfluidized nanoemulsion.
  • the oxidizing environment prevents bacterial growth.
  • the oxidizing environment is bacteriocidal.
  • the oxidizing environment provides a sterile nanoemulsion.
  • An oxidizing nanoemulsion environment may result from an increased surface to volume ratio.
  • the present invention also contemplates a method to avoid the generation of an oxidizing environment by microfluidizing in the presence of an antioxidant.
  • the antioxidant reduces the presence of reactive oxygen species (ROS) in the microfluidized nanoemulsion.
  • ROS reactive oxygen species
  • Sample Sample Sample Formulation 1 2 3 Mean Plasma 4.0 2.8 3.3 3.4 (unoxidized control) Plasma + FeCl 3 12.4 16.0 13.1 13.9 (oxidized control) 1.5 g DHA with 200 ml milk 44.0 42.6 45.8 44.1 (microfluidized) 1.75 g DHA, 1000 mg Vit E 12.8 19.2 20.1 17.4 and 800 mg Vit C with 200 ml milk (not microfluidized) 1.75 g DHA, 1000 mg Vit E 4.0 6.0 3.5 4.5 and 800 mg Vit C with 200 ml milk (microfluidized) 1.75 g DHA and 800 mg Vit C 17.5 17.8 20.8 18.7 with 200 ml milk (microfluidized) 1.75 g DHA and 1000 mg Vit E 9.8 16.7 11.4 12.6 with 200 ml milk (microfluidized) antioxidants are encapsulated by the nanoparticles for subsequent release to the subject.
  • the ROS load within any nanoemulsion preparation can be quantitatively determined by measuring indicators of an oxidizing environment.
  • Malondialdehyde (MDA) is a known indicator of an oxidizing environment.
  • This example presents one plant sterol embodiment of a microfluidized nanoemulsion.
  • the step-wise procedure is as follows:
  • step 4 Add step 4 mixture to step 5 mixture, keep stir bar and heat on for 30 mins
  • the mean particle diameter (i.e., Peak 1/Peak 2) for these microfluidized plant sterol nanoemulsions was 39 nm. See FIG. 1 .
  • the average particle diameter data for the plant sterol microfluidized nanoemulsion is shown in Table 2 below.
  • the mean particle diameter (i.e., Peak 1) for this microfluidized plant sterol nanoemulsion was 64.4 nm. See FIG. 1A .
  • the average particle diameter data for the three month plant sterol nanoemulsion is shown in Table 3 below.
  • This example presents one cod liver oil embodiment of a microfluidized nanoemulsion that has a stable particle diameter for at least four months.
  • the step-wise procedure is as follows:
  • the mean particle diameter (i.e., Peak 1/Peak 2) for this cod liver oil microfluidized nanoemulsion was 58 nm. Before microfluidization, the mean particle diameter of the cod liver oil suspension was 2,842 nm. This represents a 50-fold reduction with a single pass through the microfluidizer. Four months after the microfluidization process, the particle diameter was again determined and found not to have changed. See FIG. 2 . The average particle diameter data from the four-month microfluidized sample is presented in Table 4.
  • This example presents one tocopherol embodiment of a microfluidized nanoemulsion that maintains particle diameter for at least five months.
  • the step-wise procedure is as follows:
  • step 3 mixture to step 2 mixture
  • step 4 Add step 4 mixture to step 5 mixture, keep stir bar and heat on
  • the mean particle diameter for the tocopherol microfluidized nanoemulsion was 64 nm. Before microfluidization, the mean particle diameter for the tocopherol suspension was 1,362 nm. This represents a 21-fold reduction a single pass through the microfluidizer. Five months after the microfluidization process, the particle diameter was again determined and found not to have changed. See FIG. 3 . The average particle diameter data from the five-month microfluidized sample is presented in Table 5.
  • This example presents one lutein/zeaxanthin embodiment of a microfluidized nanoemulsion.
  • the step-wise procedure is as follows:
  • the mean particle diameter (i.e., Peak 1/Peak 2) for the lutein and zeaxanthin microfluidized nanoemulsion was 62 nm. See FIG. 4 .
  • the average particle diameter data for the sample is shown in Table 6.
  • This example presents one soy protein embodiment of a microfluidized nanoemulsion.
  • the step-wise procedure is as follows:
  • the mean particle diameter (i.e., Peak 1/Peak 2) for the vanilla soy protein (Central Soya) microfluidized nanoemulsion was 55 nm. See FIG. 5 .
  • the average particle diameter data for the sample is shown in Table 7.
  • This example presents one whey protein embodiment of a microfluidized nanoemulsion.
  • the step-wise procedure is as follows:
  • the mean particle diameter (i.e., Peak 1/Peak 2) for the whey protein microfluidized nanoemulsion was 108 nm. See FIG. 6 .
  • the average particle diameter data for the sample is shown in Table 8.
  • This example presents one orange juice/plant sterol/lutein embodiment of a microfluidized nanoemulsion.
  • the step-wise procedure is as follows:
  • the mean particle diameter (i.e., Peak 1/Peak 2) for the orange juice/plant sterol/lutein microfluidized nanoemulsion was 46 nm. See FIG. 7 .
  • the average particle diameter data for the sample is shown in Table 9.
  • This example presents one DHA fish oil/water embodiment of a microfluidized nanoemulsions that maintains particle diameter for at least two months.
  • the step-wise procedure is as follows:
  • the mean particle diameter (i.e., Peak 1) for the DHA fish oil/water microfluidized nanoemulsion was 73 nm. Two months after the microfluidization process, the particle diameter was again determined and found not to have changed. See FIG. 8 .
  • the average particle diameter data from the two-month microfluidized sample is presented in Table 10.
  • This example presents one DNA fish oil/milk embodiment without any added emulsifiers that maintains particle diameter for at least three (3) weeks.
  • the step-wise procedure is as follows:
  • the average particle diameter data from the three-week microfluidized sample is presented in Table 11.
  • This example presents the stability of microfluidized nanoemulsions following exposure to either heat or cold.
  • the formulation used in this experiment comprised DHA Fish Oil milk/tocopherol.
  • the mean particle diameter (i.e., Peak 1) for the DHA fish oil/milk/tocopherol microfluidized nanoemulsion was 87 nm. See FIG. 10 . This nano-emulsion preparation was made without any added emulsifiers. The average particle diameter data for the original nanoemulsion is presented in Table 12.
  • This original microfluidized nanoemulsion was pasteurized at 75° C. for 30 seconds. Twenty-four hours later, the oil was still in solution and the particle diameter was stable as compared to the original nanoemulsion. See FIG. 11 .
  • the average particle diameter data for the pasteurized microfluidized nanoemulsion is presented in Table 13.
  • the original microfluidized nanoemulsion was freeze-thaw tested at ⁇ 4° C. for 24 hours. Twenty-four hours later, the oil was still in solution and the particle diameter was stable as compared to the original nanoemulsion. See FIG. 12 .
  • the average particle diameter data for the freeze-thaw microfluidized nanoemulsion is presented in Table 14.
  • This example demonstrates an improved bioavailability of lycopene when fed as a uniform microfluidized nanoemulsion versus mixed into a standard diet formulation.
  • the lycopene microfluidized nanoemulsion was prepared in a step-wise manner as follows:
  • the mean particle diameter for the lycopene microfluidized nanoemulsion was 74 nm.
  • microfluidized nanoemulsion was incorporated into a chow-based diet and fed to gerbils over a 4 week period.
  • a control group was fed a lycopene in oil-enriched chow-based diet.
  • blood was collected, plasma harvested and plasma lycopene levels were determined by HPLC in both gerbil groups.
  • FIG. 13 demonstrates that control gerbils did not demonstrate detectable plasma lycopene levels.
  • a microfluidized lycopene nanoemulsion was then prepared with grape juice instead of water and orally administered to two (2) human subjects over a 4 day period (125 mg/serving, 2 servings per day). This administration raised plasma lycopene levels by approximately 38% (data not shown).
  • This example presenting data showing that microfluidized nanoemulsions provide improved efficacy over that seen in traditional nanoemulsions. Specifically, this example compares the ability of three plant sterol formulations to reduce plasma low density lipoprotein cholesterol (LDL-C) levels in hypercholesterolemic hamsters.
  • LDL-C low density lipoprotein cholesterol
  • a microfluidized mixed plant sterol (60% sitosterol) nanoemulsion was prepared in a step-wise manner as follows:
  • the mean particle diameter for the microfluidized plant sterol nanoemulsion was 41.95 nm. See FIG. 16 .
  • Group I was fed a control hypercholesterolemic diet (HCD);
  • Group 2 was fed 30 mg/d of crystalline plant sterol;
  • Group 3 was fed 20 mg/d of MinuteMaid Heartwise® micronized plant sterol (Cargill);
  • Group 4 was fed 10 mg/d of the microfluidized plant sterol nanoemulsion.
  • blood samples were analyzed for plasma LDL-C levels.
  • the microfluidized plant sterol nanoemulsion was twice as effective as the MinuteMaid Heartwise® micronized diet, and three times as effective as the crystalline plant sterol diet. See FIG. 17 .
  • Example 11 results in improved clinical therapy when compared to micron-sized or crystalline plant sterol diets.
  • This example presents data demonstrating that uniform microfluidized nanoemulsion compositions depend upon a compound having substantial solubility in the liquid dispersion medium.
  • This example compares the microfluidizing technique described in U.S. Pat. No. 5,510,118 to one embodiment as contemplated by the present invention.
  • the absorbable lipid cholesterol was chosen as the test compound.
  • Group I represents the '118 premix and was prepared by dispersing cholesterol (2 gms), water (100 mls) and Tween® 80 (0.2 gms), where cholesterol is insoluble (i.e., below at least 30 mg/ml) in the liquid dispersion medium (water). Thereafter, this cholesterol/water/Tween® 80 solution was microfluidized using a M-100EH unit. Multiple passes (10-15) through the microfluidizer were performed at PSI's ranging between 4,000-20,000 but were terminated because the generated heat exceeded 70° C. (much higher than the recommended 30-40° C. in the '118 patent. After the microfluidization it was observed that much of the cholesterol had precipitated. After twenty-four hours, the preparation of the Group I nanoemulsion contained only 0.44 gms (i.e., 22%) of the original cholesterol weight.
  • Group II represents one embodiment of the present invention and was prepared by dispersing cholesterol (2 gms) in heated soybean oil (10 gms), soy lecithin (5 gms), and Tween® 80 (0.2 gms) where cholesterol is substantially soluble (i.e., above at least 30 mg/ml) in the dispersion medium (oil). Thereafter, this cholesterol/oil/lecithin/Tween® 80 was added to 100 ml of heated water and microfluidized using a single 30 second pass at 25,000 PSI using a M-100EH unit. After the microfluidization cholesterol precipitation was not noticeably evident. After twenty-four hours, the preparation of the Group II nanoemulsion contained 1.66 gm (i.e., 83%) of the original cholesterol weight.
  • Group I shows two vastly disparate and distinct peaks. See Table 17.
  • Group II shows a single peak representing one embodiment of a uniform microfluidized nanoemulsion. See Table 18.
  • Nanoparticulate Compositions vs Uniform Microfluidized Nanoemulsions
  • This example describes a demonstration that will show that a milled nanoparticle composition (for example, one made according to US Appln Publ No. 2004/0033202 to Cooper et al.) does not create a uniform particle diameter distribution as does a microfluidized nanoemulsion as contemplated by one embodiment of the present invention.
  • An absorbable phytosterol will be chosen as the test compound.
  • Group I represents the '202 premix that will be prepared by dispersing 5% (w/w) phytosterol/water solution with 1% (w/w) Tween® 80, where the phytocholesterol is insoluble (i.e., below at least 30 mg/ml) in the liquid dispersion medium (water). Thereafter, this phytosterol/water/Tween® 80 solution will be milled at 10° C. for 1.5 to 2 hours in a DYNO®-Mill KDL (Willy A Bachofen A G, Machinefabrik, Basel, Switzerland) using a 500 ⁇ m milling media (i.e., grinding beads) of type Polymill® 500. After the milling it will be observed that much of the phytocholesterol has precipitated. After at least twenty-four hours, the preparation of the Group I nanoparticulate will contain less than 1 ⁇ 2 of the original phytosterol weight.
  • Group II represents one embodiment of the present invention and will be prepared by dispersing 5% (w/w) phytosterol/heated soybean oil solution, soy lecithin, with 1% Tween® 80, where the phytosterol is substantially soluble (i.e., above at least 30 mg/ml) in the liquid dispersion medium (oil). Thereafter, this phytosterol/oil/lecithin/Tween® 80 premix is added to 100 ml heated water and microfluidized using a single 30 second pass at 25,000 PSI using a M-100EH unit. After the microfluidization phytosterol precipitation will not be noticeably evident. After twenty-four hours, the preparation of the Group II nanoemulsion will contain greater than 3 ⁇ 4 of the original phytosterol weight.
  • Group I will most likely show at least two vastly disparate and distinct peaks. See Table 21.
  • Group II will have only a single peak representing one embodiment of a uniform microfluidized nanoemulsion. See Table 22.
  • This example will provide data showing that a uniform microfluidized nanoemulsion as contemplated by one embodiment of the present invention has improved plant sterol bioavailability and/or efficacy than a conventional nanoparticulate composition.
  • a standard curve will be constructed by gavaging thirty (30) hamsters with 1 ⁇ Ci 3 H-cholesterol. Plasma cholesterol levels are then determined at Day 1, Day 2, Day 4, and Day 7. These data are used to calculate bioavailability of 3 H-cholesterol during the 7 Day period as area-under-the-curve (AUC).
  • AUC area-under-the-curve
  • the AUC measurement will determine the ability of each preparation to reduce the absorption of 3 H-cholesterol into the bloodstream which is proportional to the bioavailability and/or efficacy of each preparation.
  • a greater bioavailability and/or efficacy of a plant sterol when administered as a uniform microfluidized nanoemulsion will be seen because: i) the average particle diameter of the uniform microfluidized nanoemulsion is smaller than the conventional nanoparticulate composition (i.e., for example, 300 nm v. 50 nm); ii) microfluidization produces more stable particles than either milling or homogenization; and iii) microfluidization produces pH-resistant particles (i.e., stomach acid or small intestine base conditions) unlike those produced by either milling or homogenization.
  • the conventional nanoparticulate composition i.e., for example, 300 nm v. 50 nm
  • microfluidization produces more stable particles than either milling or homogenization
  • iii) microfluidization produces pH-resistant particles (i.e., stomach acid or small intestine base conditions) unlike those produced by either milling or homogenization.
  • This example will provide data showing that a uniform microfluidized nanoemulsion as contemplated by one embodiment of the present invention has improved efficacy in lowering plasma cholesterol levels that a conventional nanoparticulate composition.
  • a nanoparticulate composition i.e., for example, prepared as per the '202 Cooper et al. application
  • a uniform microfluidized nanoemulsion as contemplated by one embodiment of the present invention, for four (4) additional weeks.
  • Plasma samples are taken at 0, 2, 3, 4, 5, and 6 weeks where plasma cholesterol levels will be determined by methods known in the art.
  • a greater efficacy of the plant sterol uniform microfluidized nanoemulsions to lower plasma cholesterol levels is seen because: i) the average particle diameter of the uniform microfluidized nanoemulsion is smaller than the conventional nanoparticulate composition (i.e., for example, 300 nm v. 50 nm); ii) microfluidization produces more stable particles than either milling or homogenization; and iii) microfluidization produces pH-resistant particles (i.e., stomach acid or small intestine base conditions) unlike those produced by either milling or homogenization.
  • the conventional nanoparticulate composition i.e., for example, 300 nm v. 50 nm
  • microfluidization produces more stable particles than either milling or homogenization
  • iii) microfluidization produces pH-resistant particles (i.e., stomach acid or small intestine base conditions) unlike those produced by either milling or homogenization.
  • This example provides data showing that the Bosch technique does not produce a uniform microfluidized nanoemulsion when compared to one embodiment of the present invention under identical microfluidization techniques.
  • the Group I & II premixes were prepared in accordance with Example 13. Each premix was subjected to one pass at 25,000 PSI in the microfluidizer.
  • Group I (representing the Bosch formulation) shows that 85% of the particles have a mean diameter of 815 nm. See FIG. 20A .
  • Group II (representing one embodiment of the present invention) shows that 98% of the particles have a mean diameter of 78 nm, See FIG. 20B This represents a greater than ten-fold difference in average diameter.
  • Significantly, only 15% of the Bosch particles are within the 100 nm range, thereby representing a six-fold difference in particle diameter distribution in this lower range.

Abstract

The present invention discloses an improved nanoemulsion comprising a uniform and discrete range of very small particle nano-sized diameters. This uniformity results in improved bioavailability of incorporated compounds (i.e., pharmaceuticals or nutraceuticals) as reflected in various pharmacokinetic parameters including, but not limited to, decreased Tmax, increased Cmax, and increased AUC. The improved method of making these uniform nanoemulsions utilizes microfluidization which differs in both process and mechanics when compared to conventional milling and grinding techniques used to generate nanoparticulate compositions. Further, the improvement results, in part, from a novel step of mixing a substantially soluble compound into a heated dispersion medium. This is unlike current nanoparticulate composition methods that mix an insoluble compound with an unheated dispersion medium. Further, these nanoemulsions are observed to be bacterial-resistant and stable to extremes in both temperature and pH changes. Consequently, these nanoemulsions are expected to have a significantly prolonged shelf-life than currently available nanoemulsions.

Description

    FIELD OF INVENTION
  • The present invention relates to the field of nanoemulsions. In one embodiment, nanoemulsions are made using high shear stress technology. In one embodiment, the invention comprises uniform microfluidized nanoemulsions. In another embodiment, the uniform nanoemulsion comprises a compound such as a pharmaceutical, nutraceutical, or cosmeceutical. In one embodiment, the uniform nanoemulsion comprises improved pharmacokinetic parameters when compared to conventional nanoparticulate compositions and/or nanoemulsions. In one embodiment, the present invention contemplates a method of making a bacteria-resistant nanoemulsion.
  • BACKGROUND OF THE INVENTION
  • Micro/nanoemulsion technology has substantial commercial value. In relation to the nutraceutical area alone, the market value is estimated as a 250 billion dollar business world-wide. Consequently, the ability to incorporate lipid soluble nutraceuticals into beverages (the fastest-growing component of the food industry) as well as low or no fat foods is of important interest.
  • What is needed is a nanoemulsion that: i) has improved temperature and pH stability; ii) improved bioavailability; and iii) improved shelf-life due to microbial resistance. In addition, nanoemulsions should be relatively easy and inexpensive to prepare.
  • SUMMARY
  • The present invention relates to the field of nanoemulsions. In one embodiment, the nanoemulsion is made using a high shear stress technology. In one embodiment, the invention comprises uniform microfluidized nanoemulsions. In another embodiment, the uniform nanoemulsion comprises a compound such as a pharmaceutical, nutraceutical, or cosmeceutical. In one embodiment, the uniform nanoemulsion comprises improved pharmacokinetic parameters when compared to conventional nanoparticulate compositions and/or nanoemulsions. In one embodiment, the present invention contemplates a method of making a bacteria-resistant nanoemulsion.
  • In one embodiment, the present invention contemplates a nanoemulsion comprising a population of particles having maximum and minimum diameters, wherein the difference between said maximum and minimum diameters does not exceed 100 nm.
  • In one embodiment, the present invention contemplates a nanoemulsion comprising a population of particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers. In one embodiment, the particles encapsulate a compound. In one embodiment, the compound is a pharmaceutical. In another embodiment, the compound is a nutraceutical.
  • In one embodiment, the present invention contemplates a nanoemulsion comprising a first and second population of particles, wherein the majority of particles in said first population have diameters between approximately 10 and approximately 20 nanometers, wherein the majority of particles in said second population have diameters between approximately 40 and approximately 80 nanometers, wherein said nanoemulsion is uncontaminated by particles having diameters larger than 110 nanometers. In one embodiment, the particles encapsulate a compound. In one embodiment, the compound is a pharmaceutical. In one embodiment, the compound is a nutraceutical. A nanoemulsion comprising a population of particles having diameters between approximately 50 and approximately 150 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 160 nanometers. In one embodiment, the particles encapsulate a compound. In one embodiment, the compound is a pharmaceutical. In one embodiment, the compound is a nutraceutical.
  • In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a premix comprising a compound and a liquid dispersion medium, wherein said compound has a solubility greater than 30 mg/ml in said medium; and ii) a microfluidizer capable of maintaining at least 25,000 PSI; b) using a single pass exposure of said premix to said microfluidizer to create a population of nanoemulsion particles having diameters ranging approximately between 10-110 nm. In one embodiment, the dispersion medium is selected from the group consisting of aqueous media and oil-based media. In one embodiment, the aqueous media is selected from the group consisting of water, ringers solution, dextrose, and short chain alcohols. In one embodiment, the oil-based media is selected from the group including, but not limited to, saturated and unsaturated oils from vegetable and marine sources, silicone oils, mineral oils, and plant-derived oils. In one embodiment, the compound is selected from the group including, but not limited to, a plant sterol, cod liver oil, tocopherol, lecithin, lutein, zeaxanthin, and soy protein.
  • In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a heated dispersion medium; ii) a compound having substantial solubility in said medium; and iii) a microfluidizer capable of making a uniform nanoemulsion from said medium; b) adding said compound to said medium at a temperature of at least 70° C. to create a premix; and c) microfluidizing said premix at a pressure of at least 25,000 PSI to create said nanoemulsion having particle diameters ranging between 10-110 nm. In one embodiment, said dispersion medium is selected from the group consisting of soybean oil and water. In one embodiment, said dispersion medium is heated to at least 65° C. In one embodiment, said compound may be selected from the group comprising a plant sterol, cod liver oil, tocopherol, lecithin, lutein, zeaxanthin, lycopene, whey protein, and soy protein. In one embodiment, the nanoemulsion encapsulates the compound. In one embodiment, 86% of said particle diameters have a 54 nm average diameter. In one embodiment, 14% of said particles diameters have a 16 nm average diameter. In one embodiment, 82% of said particle diameters have a 64 nm average diameter. In one embodiment, 17% of said particle diameters have a 19 nm average diameter. In one embodiment, 78% of said particle diameters have a 88 nm average diameter. In one embodiment, 22% of said particle diameters have a 27 nm average diameter. In one embodiment, 84% of said particle diameters have a 90 nm average diameter. In one embodiment, 16% of said particle diameters have a 23 nm average diameter. In one embodiment, 80% of said particle diameters have a 55 nm average diameter.
  • In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a premix comprising a compound, a first antioxidant, a second antioxidant, and an aqueous dispersion medium, wherein said compound has a solubility greater than 30 mg/ml in said medium; and iii) a microfluidizer capable of maintaining at least 25,000 PSI; c) using a single pass exposure of said premix to said microfluidizer to create a population of nanoemulsion particles having diameters ranging from between approximately 40-110 nm, wherein said particle diameter remains stable for at least four months. In one embodiment, the method further comprises pasteurizing said population of nanoemulsion particles wherein said particle diameters remain stable. In one embodiment, the method further comprises freezing said population of nanoemulsion particles wherein said particle diameters remain stable.
  • In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a stable aqueous dispersion medium comprising a first antioxidant; ii) a solution comprising natural emulsifiers; ii) a compound having substantial solubility in said medium comprising a second antioxidant; and iii) a microfluidizer capable of making a uniform nanoemulsion from said medium; b) adding said compound and said solution to said medium and heating to a temperature of at least 50° C. to create a premix; and c) microfluidizing said premix at a pressure of at least 25,000 PSI to create said nanoemulsion having particle diameters ranging between 40-110 nm wherein said particle diameter remains stable for at least four months. In one embodiment, the nanoemulsion encapsulates the compound. In one embodiment, the method further comprises pasteurizing said nanoemulsion wherein said particle diameters remain stable. In one embodiment, the method further comprises freezing said nanoemulsion wherein said particle diameters remain stable. In one embodiment, said solution comprises milk. In one embodiment, said compound comprises DHA fish oil. In one embodiment, said pasteurization comprises exposing said nanoemulsions to 75° C. for thirty seconds. In one embodiment, said freezing comprises exposing said nanoemulsions to −4° C. for 24 hours.
  • In one embodiment, the present invention contemplates a method, comprising; a) providing; i) a subject refractory to an administered compound at a therapeutically effective amount; ii) a nanoemulsion comprising a population of particles encapsulating said compound, wherein said particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers; b) delivering said nanoemulsion to said patients under conditions such that said compound bioavailability is improved and wherein said compound is therapeutically effective. In one embodiment, the improved bioavailability comprises pharmacokinetic parameters selected from the group consisting of decreased Tmax, increased Cmax, and increased AUC. In one embodiment, the delivering comprises a method selected from the group consisting of oral, transdermal, intravenous, intraperitoneal, intramuscular, and subcutaneous. In one embodiment, the nanoemulsion comprises a plant sterol. In one embodiment, the nanoemulsion comprises lycopene.
  • In one embodiment, the present invention contemplates a method for improving a nanoemulsion bioavailability comprising providing a uniform microfluidized nanoemulsion and delivering the uniform nanoemulsion to a subject. In one embodiment, the subject comprises a mammal. In one embodiment, the nanoemulsion encapsulates a compound. In one embodiment, the nanoemulsion is delivered by oral administration. In another embodiment, the nanoemulsion is delivered by methods including, but not limited to, transdermally, intravenously, intraperitoneally, intramuscularly or subcutaneously. In one embodiment, said improved bioavailability comprises pharmacokinetic parameters selected from the group consisting of decreased Tmax, increased Cmax, and increased AUC. In one embodiment, said nanoemulsion is formulated for oral administration. In one embodiment, said nanoemulsion comprises a plant sterol. In one embodiment, said nanoemulsion comprises lycopene.
  • In one embodiment, the present invention contemplates a nanoemulsion having bacteria-resistant properties, wherein said nanoemulsion comprises a population of particles encapsulating said compound, wherein said particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers. In one embodiment, the nanoemulsion resists bacterial growth for at least three months. In one embodiment, the bacterial-resistant properties comprise shear-force induced cell lysis. In one embodiment, the bacterial-resistant properties comprise an oxidizing environment. In one embodiment, the nanoemulsion is sterile.
  • In one embodiment, the present invention contemplates a uniform microfluidized nanoemulsion comprising bacteria-resistant properties. In one embodiment, said nanoemulsion resists bacterial growth for at least three months. In one embodiment, the nanoemulsion comprises particles having a diameter distribution of between 10-110 nm. In one embodiment, said bacterial-resistant properties comprise shear-force induced cell lysis. In one embodiment, the nanoemulsion is sterile.
  • In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a premix comprising a compound and a liquid dispersion medium; and ii) a device capable of creating a continuous turbulent flow under high pressure; b) using said device to create a population of nanoemulsion particles having uniform diameter. In one embodiment, the dispersion medium is selected from the group consisting of aqueous media and oil-based media. In one embodiment, the aqueous media is selected from the group consisting of water, saline solution, ringers solution, dextrose, and short chain alcohols. In one embodiment, the oil-based media is selected from the group consisting of saturated and unsaturated oils from vegetable and marine sources, silicone oils, and mineral oils. In one embodiment, the compound is selected from the group consisting of a plant sterol, cod liver oil, tocopherol, lecithin, lutein, zeaxanthin, and soy protein.
  • DEFINITIONS
  • In general, the terms used herein are to be interpreted according to definitions generally accepted by those having ordinary skill in the art. Those listed below, however, are to be interpreted according to the following definitions.
  • The term “microfluidized”, “microfluidizing”, or “microfluidizer” as used herein refers to an instrument or a process that utilizes a continuous turbulent flow at high pressure including, but not limited to, a microfluidizer or other like device that may be useful in creating a uniform nanoemulsion. For example, microfluidizing may create a uniform nanoemulsion comprising a pharmaceutical, nutraceutical, or cosmeceutical from a premix within a thirty (30) second time frame (typically referred to a single pass exposure). Typically, a microfluidizer may be operated at a pressure of approximately 25,000 PSI to generate a uniform nanoemulsion.
  • The term “uniform nanoemulsion” as used herein, refers to any emulsion comprising any specified range of particle diameter sizes wherein the difference between the minimum diameter and maximum diameters do not exceed approximately 600 nm, preferably approximately 300 nm, more preferably approximately 200 nm, but most preferably approximately 100 nm (i.e., for example, microfluidization, as contemplated herein, produces a uniform nanoemulsion having a range of approximately 10-110 nm and is referred to herein as a uniform microfluidized nanoemulsion). Preferably, the total particle distribution (i.e., 100%) is encompassed within the specified range of particle diameter size. A particle diameter distribution where less than 3% is outside the specified range of particle diameter sizes is still contemplated herein as a uniform nanoemulsion.
  • The term “population” as used herein, refers to any mixture of nanoemulsion particles having a distribution in diameter size. For example, a population of nanoemulsion particles may range is particle diameter from between approximately 10-10 nm.
  • The term “nanoparticle” as used herein, refers to any particle having a diameter of less than 300 nanometers (nm), as defined by the National Science Foundation or preferably less than 100 nm, as defined by the National Institutes of Health. Most conventional techniques create nanoparticle compositions with an average particle diameter of approximately 300 nanometers (nm) or greater.
  • The term “dispersion medium” as used herein, refers to any oil-based or aqueous liquid wherein a pharmaceutical, nutraceutical, or cosmeceutical may be dissolved upon heating. Oil-based liquids may include, but not limited to; saturated and unsaturated oils from vegetable and marine sources including, but not limited to, soybeans, safflowers, olives, corn, cottonseeds, linseed, safflower, palm, peanuts, flaxseeds, sunflowers, rice bran, sesame, rapeseed, cocoa butter etc., and mixtures thereof; silicone oils; and mineral oils. Alternatively, aqueous media may include, but are not limited to, water, saline solutions, short chain alcohols, 5% dextrose, Ringer's solutions (lactated Ringer's injection, lactated Ringer's plus 5% dextrose injection, acylated Ringer's injection), Normosol-M, Isolyte E, and the like; and synthetic and/or natural detergents having high surfactant properties, deoxycholates, cyclodextrins, chaotropic salts and ion pairing agents etc., and mixtures thereof.
  • The term “compound” as used herein, refers to any pharmaceutical, nutraceutical, or cosmeceutical (i.e., for example, organic chemicals, lipids, proteins, oils, vitamins, crystals, minerals etc.) that are substantially soluble in a dispersion medium.
  • The term “substantially soluble” as used herein, refers to any compound that dissolves into a dispersion medium to a concentration greater than 30 mg/ml. Preferably, the dispersion medium is heated while the compound is being dissolved.
  • The term “premix” as used herein, refers to any mixture that is subsequently used to generate a nanoparticulate composition or a uniform microfluidized nanoemulsion. Typically, premixes contain a liquid dispersion medium and a compound, and optionally, an emulsifier and/or an antioxidant.
  • The term “stable” as used herein, refers to any population of nanoemulsion particles whose diameters stay within the range of approximately 10-110 nm over a prolonged period of time (i.e., for example, one (1) day to twenty-four (24) months, preferably, two (2) weeks to twelve (12) months, but more preferably two (2) months to five (5) months). For example, if a population of nanoemulsion particles is subjected to prolonged storage, temperature changes, and/or pH changes whose diameters stay within a range of between approximately 10-110 nm, the nanoemulsion is stable.
  • The term “bacteria-resistant” as used herein refers to the lack of observable bacterial growth.
  • The term “sterile” as used herein refers to a nanoemulsion that contains no living bacterial cells.
  • The term “pharmaceutically acceptable” as used herein, refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • The term “pharmaceutically acceptable salts” as used herein, refers to derivatives wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • The term “therapeutically effective amount” as used herein, with respect to a drug dosage, shall mean that dosage that provides the specific pharmacological response for which the drug is administered or delivered to a significant number of subjects in need of such treatment. It is emphasized that ‘therapeutically effective amount,’ administered to a particular subject in a particular instance will not always be effective in treating the diseases described herein, even though such dosage is deemed a “therapeutically effective amount” by those skilled in the art. Specific subjects may, in fact, be “refractory” to a “therapeutically effective amount”. For example, a refractory subject may have a low bioavailability such that clinical efficacy is not obtainable. It is to be further understood that drug dosages are, in particular instances, measured as oral dosages, or with reference to drug levels as measured in blood.
  • The term “refractory” as used herein, refers to any subject that does not respond with an expected clinical efficacy following the delivery of a compound as normally observed by practicing medical personnel.
  • The term “delivering” or “administering” as used herein, refers to any route for providing a pharmaceutical or a nutraceutical to a subject as accepted as standard by the medical community. For example, the present invention contemplates routes of delivering or administering that include, but are not limited to, oral, transdermal, intravenous, intraperitoneal, intramuscular, or subcutaneous.
  • The term “subject” as used herein, refers to any animal to which an embodiment of the present invention may be delivered or administered. For example, a subject may be a human, dog, cat, cow, pig, horse, mouse, rat, gerbil, hamster etc.
  • The term “encapsulate”, “encapsulated”, or “encapsulating” refers to any compound that is completely surrounded by a protective material. For example, a compound may become encapsulated by a population of nanoemulsion particle formation during microfluidization.
  • The term “nutraceutical” refers to any compound added to a dietary source (i.e., for example, a fortified food or a dietary supplement) that provides health or medical benefits in addition to its basic nutritional value.
  • The term “cosmeceutical” refers to any compound (i.e., for example, benzoyl peroxide or retinol) added to a preparation that possesses both cosmetic and pharmaceutical properties. A cosmecuetical is generally useful for external applications to improve the complexion or overall physical appearance. Cosmeceuticals may be applied as compositions including, but not limited to, a cream, oil, foam, spray, liquid etc. Cosmeceuticals may include categories such as, but not limited to, carotenoids, phenolic compounds, or water soluble antioxidants.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 presents exemplary data showing the particle diameter distribution of a microfluidized plant sterol nanoemulsion population three (3) months after preparation.
  • FIG. 1A presents exemplary data showing the particle diameter distribution of a microfluidized plant sterol nanoemulsion three (3) months after preparation.
  • FIG. 2 presents exemplary data showing the particle diameter distribution of a microfluidized cod liver oil nanoemulsion population four (4) months after preparation.
  • FIG. 3 presents exemplary data showing the particle diameter distribution of a microfluidized tocopherol nanoemulsion population five (5) months after preparation.
  • FIG. 4 presents exemplary data showing the particle diameter distribution of a microfluidized lutein/zeaxanthin nanoemulsion population.
  • FIG. 5 presents exemplary data showing the particle diameter distribution of a microfluidized soy protein nanoemulsion population.
  • FIG. 6 presents exemplary data showing the particle diameter distribution of a microfluidized whey protein nanoemulsion population.
  • FIG. 7 presents exemplary data showing the particle diameter distribution of a microfluidized orange juice/plant sterol/lutein nanoemulsion population.
  • FIG. 8 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/water nanoemulsion population two (2) months after preparation.
  • FIG. 9 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/milk nanoemulsion population three (3) weeks after preparation.
  • FIG. 10 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/milk/tocopherol nanoemulsion population.
  • FIG. 11 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/milk/tocopherol nanoemulsion population after pasteurization.
  • FIG. 12 presents exemplary data showing the particle diameter distribution of a microfluidized DHA fish oil/milk/tocopherol nanoemulsion population after a freeze-thaw process.
  • FIG. 13 presents exemplary data of gerbil plasma lycopene levels when fed a lycopene-enriched diet.
  • FIG. 14 presents exemplary data of gerbil plasma lycopene levels when fed a microfluidized lycopene nanoemulsion diet.
  • FIG. 15 presents one embodiment of an anti-bacterial property generated during the preparation of a microfluidized plant sterol nanoemulsion.
  • FIG. 16 presents exemplary data showing the particle diameter distribution of a microfluidized plant sterol nanoemulsion population used in Example 12.
  • FIG. 17 presents exemplary data showing that a microfluidized plant sterol nanoemulsion diet is more effective in reducing plasma LDL-C in hypercholesterolemic hamsters than either a micronized plant sterol diet or a crystalline plant sterol diet for four (4) weeks.
  • FIG. 18 presents exemplary data comparing premix cholesterol particle diameter distributions from: Panel A: Tween® 80/Water as per the '118 patent; and Panel B: Oil/Lecithin/Tween® 80/Water as contemplated by one embodiment of the present invention.
  • FIG. 19 presents exemplary data comparing microfluidized cholesterol nanoemulsion particle diameter distributions from: Panel A: Tween® 80/Water as per the '118 patent using repeated microfluidization passes; and Panel B: Oil/Lecithin/Tween® 80/Water as contemplated by one embodiment of the present invention using a single microfluidization pass.
  • FIG. 20 presents exemplary data comparing microfluidized cholesterol nanoemulsion particle diameter distributions from: Panel A: Tween® 80/Water as per the '118 patent using a single pass exposure; and Panel B: Oil/Lecithin/Tween® 80/Water as contemplated by one embodiment of the present invention using a single pass exposure.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to the field of nanoemulsions. In one embodiment, the nanoemulsion is created by a high shear stress technology. In one embodiment, the invention comprises uniform microfluidized nanoemulsions. In another embodiment, the uniform nanoemulsion comprises a compound such as a pharmaceutical, nutraceutical, or cosmeceutical. In one embodiment, the uniform nanoemulsion comprises improved pharmacokinetic parameters when compared to conventional nanoparticulate compositions and/or nanoemulsions. In one embodiment, the present invention contemplates a method of making a bacteria-resistant nanoemulsion.
  • The use of nanoemulsions as a delivery system is generally directed to pharmaceuticals. Nanoemulsion nutraceutical delivery, however, has received little attention. For example, one nanoemulsion system contains plant sterols. Bruce et al., “Method for producing dispersible sterol and stanol compounds” U.S. Pat. No. 6,387,411 (2002) (herein incorporated by reference). This technology, however, uses a grinding method to produce the nanoemulsions, and consequently, the particle diameter is at least six (6) times greater than contemplated herein. Although it is not necessary to understand the mechanism of an invention, it is believed that this diameter difference offers particular advantages in stability and efficacy (infra). Further, the '411 patent does not disclose the incorporation of absorbable micronutrients.
  • A further use of nanoemulsions as a delivery system is directed to cosmeceuticals. Cosmeceuticals may comprise, for example; carotenoids including, but not limited to, α-carotene, β-carotene, β-cryptoxanthin, lycopene, crocetin, fucoxanthin, halocynthiaxanthin, canthaxanthin, astraxanthin, lutein, or zeaxanthin; phenolic compounds including, but not limited to, quercetin, rutin, myricetin, kaemferol, catechin, epigallocatechin, epicatechin, reservatrol, tocopherol, ferulate, ubiquinol-10, soy isoflavones such as genestein, daidzein, alpha lipoic acid, anthocyanins, ellagic tannins, gallic or ellagic acids; or water soluble anti-oxidants such as ascorbic acid, uric acid, or bilirubin.
  • The present invention is directed to populations of nanoparticles or nanoemulsions comprising an oral delivery vehicle for all absorbable (i.e., for example, fat-soluble) nutrients including, but not limited to, fatty acids, carotenoids, tocopherols, tocotrienols, and coenzyme-Q. Delivery methods, however, are not limited to oral and include, but are not limited to, transdermal, intravenous, intraperitoneal, intramuscular, or subcutaneous. In another embodiment, the carotenoids include, but are not limited to, lutein and zeaxanthin. The present invention is also directed to populations of nanoparticles or nanoemulsions comprising an oral delivery vehicle for all non-absorbable (i.e., for example, fat soluble) plant sterol compounds including, but not limited to, phytosterols and phytostanols. In one embodiment, the compounds are encapsulated by the nanoparticles or nanoemulsions. In one embodiment, common emulsifying agents are used to prepare the nanoemulsions. In one embodiment, the emulsifying agents include, but are not limited to, phospholipids, fatty acid monoglycerides, fatty acid diglycerides, or polysorbates.
  • The present invention also contemplates that certain nanoemulsion embodiments of the present invention comprise a surface-to-volume ratio that results in an improved bioavailability over current methods and compositions known in the art.
  • The present invention also contemplates that certain nanoemulsion embodiments of the present invention are resistant to microbiological growth. Although it is not necessary to understand the mechanism of an invention, it is believed that the microfluidization process comprises a high sheer stress and/or creates an oxidizing environment, thereby disrupting microbial integrity and/or preventing microbial growth.
  • 1. Methods of Making Nanoemulsions
  • Nanoemulsions have been generated by a variety of methods. In particular, these methods provide a wide variation in particle diameter and require organic solvents and or polymers. When these known nanoemulsions are considered for an oral drug or nutrient delivery system, issues of biocompatibility and physiological side effects become an important issue.
  • In one embodiment, the present invention contemplates a method of making a nanoemulsion comprising a continuous turbulent flow at high pressure. In one embodiment, the high pressure turbulent flow comprises microfluidization. In one embodiment, a uniform nanoemulsion is generated from a premix using a single pass exposure (i.e., for example, within a thirty (30) second time frame). In one embodiment, the uniform nanoemulsion comprises a population of particles whose difference between the minimum and maximum diameters does not exceed approximately 100 nm. In one embodiment, a uniform nanoemulsion is generated using a pressure of at least 25,000 PSI. In one embodiment, the present invention contemplates a method of making uniform microfluidized nanoemulsions without organic solvents or polymers. In one embodiment, the microfluidized nanoemulsion is made from a suspension. In another embodiment, the microfluidized nanoemulsion is made from a microemulsion.
  • In one embodiment, the present invention contemplates a uniform microfluidized nanoemulsion using compounds that are substantially soluble in a liquid dispersion medium. In one embodiment, the nanoemulsion encapsulated the compounds. In one embodiment, the compounds comprise pharmaceuticals and/or nutraceuticals.
  • Exemplary nutraceuticals and dietary supplements are disclosed, for example, in Roberts et al., Nutriceuticals: The Complete Encyclopedia of Supplements, Herbs, Vitamins, and Healing Foods (American Nutriceutical Association, 2001), which is specifically incorporated by reference. Dietary supplements and nutraceuticals are also disclosed in Physicians' Desk Reference for Nutritional Supplements, 1st Ed. (2001) and The Physicians' Desk Reference for Herbal Medicines, 1st Ed. (2001), both of which are also incorporated by reference. A nutraceutical or dietary supplement, also known as a phytochemical or functional food, is generally any one of a class of dietary supplements, vitamins, minerals, herbs, or healing foods that have medical or biological 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, amino acids (e.g., glutamine, arginine, iso-leucine, leucine, lysine, methionine, phenylalanine, 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.”
  • In particular, these compounds include, but are not limited to, naturally occurring oils, fatty acids, and proteins. In one embodiment, a naturally occurring oil comprises fish oil (i.e., for example, cod liver oil). In one embodiment, a naturally occurring fatty acid comprises an omega-3 (i.e., for example, DHA). In one embodiment, the nanoemulsion comprises little or no fat. In one embodiment, a naturally occurring protein comprises soy or whey.
  • In one embodiment, the present invention contemplates a method of making a uniform microfluidized nanoemulsion comprising a population of particles whose diameter ranges from between 10-110 nm.
  • A. The Microfluidizer
  • Microfluidization is a unique process that powers a single acting intensifier pump. The intensifier pump amplifies the hydraulic pressure to the selected level which, in turn, imparts that pressure to the product stream. As the pump travels through its pressure stroke, it drives the product at constant pressure through the interaction chamber. Within the interaction chamber are specially designed fixed-geometry microchannels through which the product stream will accelerate to high velocities, creating high shear and impact forces that generates a uniform nanoemulsion as the high velocity product stream impinges on itself and on wear-resistant surfaces.
  • As the intensifier pump completes its pressure stroke, it reverses direction and draws in a new volume of product. At the end of the intake stroke, it again reverses direction and drives the product at constant pressures, thereby repeating the process.
  • Upon exiting the interaction chamber, the product flows through an onboard heat exchanger which regulates the product to a desired temperature. At this point, the product may be recirculated through the system for further processing or directed externally to the next step in the process. Cook et al., “Apparatus For Forming Emulsions” U.S. Pat. No. 4,533,254 (1985); and Cook et al., “Method Of Forming A Microemulsion” U.S. Pat. No. 4,908,154 (1990) (both herein incorporated by reference).
  • B. Nanoparticulate Compositions
  • Early attempts using microfluidizers to create nanoparticulate compositions required drug substances that were poorly soluble in a liquid dispersion medium. In one disclosed technology, “poorly soluble” was defined as less than 10 mg/ml. Bosch et al., “Process for preparing therapeutic compositions containing nanoparticles” U.S. Pat. No. 5,510,118 (1996) (herein incorporated by reference). While water-insolubility was preferably considered, oil-insoluble compounds were also subjected to a microfluidization process. The '118 patent's microfluidization process is described as a “milling” action, thus indicating that the insoluble compound particles are undergoing a physical disintegration during the creation of the nanoparticulate composition. Further, this previous process requires long-processing times (i.e., repeated microemulsifying cycles) thereby promoting heat build-up in the microfluidizer. Consequently, this early technique requires processing temperatures of less than 40° C. One problem is that this technique resulted in average nanoemulsion particle diameters of approximately 300 nm. Despite teachings within the '118 patent that lower particle diameters (i.e., less than 100 nm) can be achieved, no data is presented demonstrating such a capability. Exemplary data presented herein has used the Bosch et al. process to produce a complete particle diameter distribution profile. See Example 13. These data show that the Bosch et al. technology cannot produce a uniform nanoemulsion as contemplated by the present invention.
  • Several others have implemented the basic '118 technology to encapsulate various insoluble compounds. In fact, these subsequent disclosures define a nanoparticle composition as “particles consisting of a poorly soluble therapeutic or diagnostic agent having adsorbed onto, or associated with, the surface thereof a non-crosslinked surface stabilizer”. Cooper et al., “Nanoparticulate Sterol Formulations And Novel Sterol Combinations” United States Patent Application Publication No. 2004/0033202 A1 (2004) (see pg 1 para 3) (herein incorporated by reference). Like the '118 patent, Cooper et al. discloses preparing nanoparticulate compositions using compounds that are poorly soluble in a liquid dispersion medium (i.e., water, oils, alcohols, glycols, etc.). For example, Cooper et al. defines a “poorly water-soluble” drug as having a solubility of less than about 30 mg/ml. For example, plant sterol nanoparticulate compositions comprising one or more sterols or stanols (i.e., sitosterol or phytosterols) are suggested in the art as having a particle diameter of less than 50 nm. Cooper et al. does not use a microfluidizer nor present any data showing a capability of providing a uniform particle diameter ranging between 10-110 nm. Instead, Cooper et al. relies upon a more traditional milling process that does not produce a uniform particle diameter distribution ranging between 10-110 nm.
  • Cooper et al. employs a milling grinder known in the art as a DYNO®-MILL KDL. This equipment is currently marketed in the United States by Glen Mills, Inc. (Clifton, N.J.) and advertises with the following technical information. The DYNO®-MILL is a versatile horizontal bead mill having applications ranging from paints and coatings to drug manufacturing and cell disruption for extracting proteins. Grinding to a mean diameter of 320 nm has been reported in research papers. Operation of the DYNO®-MILL is always wet, that is, the material to be ground is held in suspension in any suitable liquid. A jacketed grinding chamber contains a series of agitators that are equally spaced along the length of a central shaft. The jacket on the grinding chamber is used to control the temperature of the material being processed. The chamber is secured at one end and cantilevers out over the shaft. The bearing end contains a separator gap which has clearance tolerances that can be set as tight as 20 microns. The chamber is filled to about 80% of its capacity with beads (i.e., PolyMill® 500; 500 μm diameter grinding beads). Depending on the specific application beads made from glass, ceramic, metals, tungsten carbide and other materials are available. The process material is now introduced into the chamber. When the chamber is full of material and beads, the machine is switched on and the agitator discs rotate forcing the beads to impact over and over with the process material with hurricane-like force. This action of having thousands of separate impacts produces rapid and consistent size reduction. Batch and continuous processing can be handled in the same mill by changing the grinding chamber and the gap setting.
  • At best, Cooper et al. is limited to a plant sterol nanoparticulate composition where 90% of the particle diameters are below 187 nm. The actual particle diameter distribution, however, is not presented. In one embodiment, the present invention contemplates that the technology described by Cooper et al. cannot produce a uniform particle diameter distribution ranging between 10-110 nm. See Example 14. Unlike some embodiments of the present invention, Cooper et al. has not considered methods to make a nanoparticulate composition that include a heating process. In fact, Cooper et al. presents a discussion concluding that preparing a plant sterol nanoparticulate composition using a process that includes heating is not desirable and problematic. Some embodiments of the present invention have solved those problems.
  • Two drugs that are insoluble in a selected liquid dispersion medium, meloxicam and topiramate, are suggested as potential candidates for improved clinical administration using the Cooper et al. nanoparticulate composition technology. Cooper et al., “Nanoparticulate meloxicam formulations” US Pat. Appln Publ. No. 2004/0229038 (2004); and Gustow et al., “Nanoparticulate topiramate formulations” US Pat. Appln Publ. No. 2004/0258758 (2004). Neither publication contains any exemplary data demonstrating the creation of a uniform microfluidized microemulsion having a particle diameter range of about 10-110 nm.
  • C. Nanoemulsification
  • The formation of a uniform mixture (i.e., for example, a population) of predominantly small particles may involve a physical process termed “emulsification”. An emulsion is traditionally defined in the art “as a system . . . consisting of a liquid dispersed with or without an emulsifier in an immiscible liquid usually in droplets of larger than colloidal size” Medline Plus Online Medical Dictionary, Merriam Webster (2005). Consequently, as the art developed emulsifiers capable of generating smaller and smaller diameter particles, the terms “microemulsion” and “nanoemulsion” became known. Conceptually, a microemulsion is one thousand-fold greater in diameter than a nanoemulsion. However, particle diameter distributions may vary widely in a non-controlled emulsification process creating considerable overlap between the nanoemulsion and microemulsion technologies.
  • In one embodiment, the present invention contemplates a premix comprising a compound substantially soluble (i.e., for example, greater than 30 mg/ml) in a liquid dispersion medium (i.e., for example, a heated liquid dispersion medium) and, optionally, common emulsifying agents including, but not limited to, phospholipids, fatty acid monoglycerides, fatty acid diglycerides, or polysorbates. In one embodiment, a nanoemulsion is created by exposing a premix to a continuous turbulent flow at a high pressure, wherein the pressure is at least 25,000 PSI. In one embodiment, the high pressure turbulent flow comprises microfluidization. In one embodiment, the nanoemulsion comprises particles encapsulating pharmaceuticals or nutraceuticals. In one embodiment, the nanoemulsion comprises a uniform nanoemulsion having stable particles. In one embodiment, the microfluidization comprises a single pass exposure (i.e., for example, approximately thirty (30) seconds). In one embodiment, a uniform plant sterol microfluidized nanoemulsion has an improved low density lipoprotein cholesterol lowering efficacy.
  • Oral drug administration is a common method for providing pharmaceuticals and nutraceuticals to any subject. The contemplated methods of delivering a nanoemulsion is not limited to oral and include, for example, transdermal, intravenous, intraperitoneal, intramuscular, or subcutaneous routes of administration. Oral administration is favored because the formulations (i.e., liquids or suspensions) are relatively inexpensive to produce and are well tolerated. Subsequent gastrointestinal absorption of the formulation's ingredients, however, is not as predictable. For the pharmaceuticals and nutraccuticals to gain entrance into the subject, the formulations must be compatible with the digestive system. Consequently, lipid-based drug delivery systems are known to be useful as carriers for many drug delivery systems. Their efficacy, however, may be dependent upon; i) lipid composition (i.e., for example, molecular size and charge); ii) pharmaceutical, nutraceutical, or cosmeceutical chemical structure (i.e., molecular size and pH ionization); and iii) the overall health of the subject. Lipids are generally categorized as physiologically non-absorbable or absorbable. It should be recognized that gastrointestinal absorption processes are unrelated to a compound's solubility properties. The present invention contemplates compositions and methods related to uniform microfluidized nanoemulsions comprising either absorbable or non-absorbable lipids thereby improving their bioavailability.
  • 1. Non-Absorbable Lipids
  • Plant sterols, stanols, and triterpene alcohols (i.e., for example, oryzanol) are either not absorbed, or poorly absorbed, into the bloodstream following oral administration. In one embodiment, the present invention contemplates a method of making a uniform nanoemulsion (i.e., for example, microfluidized) comprising a non-absorbable lipid having substantial solubility in a liquid dispersion medium and, optionally, common emulsifying agents, such as phospholipids, fatty acid monoglycerides, fatty acid diglycerides, or polysorbates to formulate improved nanoemulsions. In one embodiment, the nanoemulsion comprises particle diameters ranging between 10-110 nm, thereby improving oral administration.
  • The use of plant sterols, such as β-sitosterol, is known to reduce blood cholesterol levels because it is non-absorbable. The presence of unabsorbed plant sterols in the gastrointestinal system inhibits the normal metabolism of cholesterol and, concomitantly, decreases blood cholesterol levels. Specifically, administration of twenty (20) gms of crystalline plant sterols can reduce plasma cholesterol levels approximately 10%. Pollack et al., “Sitosterol”, In: Monographs on Atherosclerosis, Vol. 10, Eds. O. J. Pollack & D. Kritchevsky, Basel, N.Y., Karger (1981).
  • Further, non-absorbable lipids are advantageous as a nutraceutical because of a lack of side effects. Side effects are routinely observed when using traditional pharmaceutical systemic cholesterol-lowering interventions (i.e., for example, HMG CoA reductase inhibitors or niacin). Because of the very low incidence of side effects, plant sterols can be prescribed for the general population, including children for whom systemic interventions are rarely recommended. It is known that the consumption of adequate amounts of plant sterols will lower blood cholesterol levels. The present invention contemplates improvements in currently known methods to deliver plant sterols or stanols.
  • The first known method involves dissolving the plant sterol in a vegetable oil-containing margarine to an efficacious level of plant sterol. When the fat solubility of a free stanol or a sterol is increased by: i) interesterified with a fatty acid such oleate or linoleate; ii) mixed in vegetable oil; or iii) hydrogenated to produce margarine, plasma cholesterol can be reduced by approximately 30%. To ingest enough plant sterol, this process can result in the consumption of up to approximately eighteen (18) grams of fat. Miettinen et al., “Use of a stanol fatty acid ester for reducing serum cholesterol level” U.S. Pat. No. 5,502,045 (1996); and Wester et al., “Phytosterol compositions” U.S. Pat. No. 6,589,588 (2003) (both herein incorporated by reference). To fat conscious Americans, coupled with the high cost of the margarines, this is unacceptable for a naturopathic approach to lower plasma cholesterol. A disadvantage of this method is that overweight or obese people frequently have elevated cholesterol levels. Physicians, of course, caution this subject group to avoid additional dietary fat. In one embodiment, the present invention contemplates a method of making a beverage nanoemulsion that comprises plant sterols. For example, the method to make the beverage nanoemulsion may comprise a continuous turbulent flow at a high pressure. In one embodiment, the continuous turbulent high pressure flow comprises microfluidization. In another embodiment, the nanoemulsion beverage comprises an orange juice product.
  • The second known method comprises oral delivery of water-dispersible plant sterols (i.e., for example, a stanol not dissolved in fat) by incorporation micron-sized micelles (i.e., microemulsions having diameters of several thousand nanometers) which can be subsequently added to beverages or foods. Ostlund, Jr., “Sitostanol formulation to reduce cholesterol absorption and method for preparing and use of same” U.S. Pat. No. 5,932,562 (1999)(herein incorporated by reference). When the microemulsion containing the plant sterol was administered into the intestine, cholesterol absorption was reduced by approximately 37%. Ostlund, Jr., “Sitostanol formulation to reduce cholesterol absorption and method for preparing and use of same” U.S. Pat. No. 5,932,562 (1999)(herein incorporated by reference); and Spillburg et al., “Fat-free foods supplemented with soy stanol-lecithin powder reduce cholesterol absorption and LDL cholesterol” J Am Diet Assoc. 103:577-581 (2003). A disadvantage of this method is that the particle diameters of these microemulsion preparations are on the order of thousands of nanometers (i.e., micron diameters) and thereby does not provide optimal efficacy. The present invention contemplates a nanoemulsion technology comprising a specific formulation and a microfluidization process that provides particle diameters from between 10-110 nm. In one embodiment, the nanoparticle has improved pH and temperature stability properties, thereby stabilizing the particle's integrity throughout the gastrointestinal system.
  • The third known method involves the oral delivery of plant sterols by producing a water dispersible sterol product. These water dispersible products usually include emulsifying agents including, but not limited to, monoglycerides and polysorbates. These water dispersible products are known to be homogenized using a liquid/liquid dispersion having particle diameters less than 1000 nm (mean=358 nm). The present invention, however, contemplates a microfluidizing nanoemulsion technology (i.e., for example, that produced by a continuous flow high pressure process) that improves the emulsification of these water-dispersible plant sterols into nanoemulsions having a particle diameter of approximately 40-60 nm.
  • Similarly, methods are known for preparing water dispersible sterol/stanol or sterol/stanol ester compositions by co-melting the stanol/sterols with highly branched hydrocarbons and then grinding the resulting product. Bruce et al., “Method for producing dispersible sterol and stanol compounds” U.S. Pat. No. 6,387,411 (2002) (herein incorporated by reference). This grinding method typically produces particle diameters ranging from 10-150 microns. Other methods known to produce a water dispersible sterol product use homogenization in emulsifying agents including, but not limited to, monoglycerides and polysorbates. These homogenization procedures have been reported to produce a liquid/liquid dispersion with a particle diameter less than 1000 nm (mean=358 nm). Stevens et al., “Aqueous dispersible sterol product” U.S. Pat. No. 6,623,780 (2003) (herein incorporated by reference). This preparation, when added to orange juice, can reduce LDL cholesterol by approximately 12%. Devaraj et al., “Plant sterol-fortified orange juice effectively lowers cholesterol levels in mildly hypercholesterolemic healthy individuals” Arterioscler Thromb Vasc Biol. 24:25-28 (2004).
  • Although it is not necessary to understand the mechanism of an invention, it is believed that a much greater surface-to-volume ratio is reached in the uniform microfluidized nanoemulsion preparations made according to the present invention (i.e., for example, up to 6 fold) and results in greater stability. Consequently, it is further believed that, any incorporated pharmaceutical, nutraceutical, or cosmeceutical has improved efficacy (i.e., for example, plasma cholesterol lowering by a plant sterol). It is further believed that a smaller-sized plant sterol-containing nanoparticles contemplated by one embodiment of the present invention, when compared to known micron-sized micelles or microemulsions, has an improved disruption of the normal micellar delivery of dietary cholesterol to the digestive tract. For example, it is known that pre-formed micron-size micelles containing plant stanols were up to three (3) times more efficacious in inhibiting cholesterol absorption than a suspension of crystalline stanol. Ostlund et al., “Sitostanol administered in lecithin micelles potently reduces cholesterol absorption in humans” Am J Clin Nutr 70:826-831 (1999).
  • 2. Absorbable Lipids
  • This invention also relates to the use of nanoemulsions as an oral delivery vehicle for absorbable lipids including, but not limited to, fatty acids, carotenoids, tocopherols and other fat soluble vitamins, tocotrienols, and Coenzyme-Q. In one embodiment, the present invention contemplates a method to make a uniform microfluidized nanoemulsion comprising an absorbable lipid having substantial solubility in a liquid dispersion medium and, optionally, common emulsifying agents, such as phospholipids, fatty acid monoglycerides, fatty acid diglycerides, or polysorbates to formulate improved nanoemulsions. In one embodiment, the method comprises a step exposing a premix to a continuous turbulent flow at high pressure. In one embodiment, the pressure is at least 25,000 PSI. In one embodiment, the nanoemulsion comprises carotenoids, including, but not limited to, lutein and zeaxanthin. In one embodiment, the nanoemulsion comprises nanoparticles having a particle diameter ranging from 10-110 nm, thereby improving bioavailability. In one embodiment, nanoemulsion bioavailability is improved following oral, transdermal, intravenous, intraperitoneal, intramuscular or subcutaneous delivery.
  • In one embodiment, the present invention contemplates a method to treat or prevent macular degeneration (i.e., a major cause of blindness in people of 65) providing an improved nanoemulsion comprising at least one carotenoid. In one embodiment, the carotenoid is selected from the group comprising lutein or zeaxanthin.
  • Under normal physiological conditions these types of compounds may be poorly absorbed by the gastrointestinal system. Consequently predicable lipid nutrient absorption is highly variable thus resulting in a highly variable lipid bioavailability (i.e., for example, the percentage of the dose absorbed). Factors influencing bioavailability may include, but are not limited to, food processing methods, food matrix, and physiological solubility in naturally-occurring micelles (i.e., for example, the lipid micellular transport system).
  • Fat-soluble nutrients can be incorporated into high fat-containing vegetable oils for dispersal into a fat matrix (i.e., for example a micron-sized micelle). The micelle solubilizes the lipid-soluble nutrient thereby allowing absorption by the small intestine. For example, when plant sterols are delivered in a micelle, cholesterol absorption inhibition is increased up to three-fold. Ostlund et al., “Sitostanol administered in lecithin micelles potently reduces cholesterol absorption in humans” Am J Clin Nutr 70:826-831 (1999).
  • Similarly, an increased in vitro carotenoid bioavailability in cell cultures is observed when solubilizing the carotenoids in micelles. Xu et al., “Solubilization and stabilization of carotenoids using micelles: delivery of lycopene to cells in culture” Lipids 34:1031-1036 (1999). A disadvantage of using micelles, however, involves the use of chlorinated organic solvents, a practice that should be avoided in the processing of foods stuffs. Another in vitro experiment demonstrates that a nanoemulsion preparation of lipophilic substances, such as fatty acids, vitamins, and beta-carotene can be delivered into cell culture medium (RPMI-1640) and incorporated by TK-6 cells. Zuelli et al., “Delivering lipophilic substances into cells using nanoemulsions” U.S. Pat. No. 6,558,941 (2003) (herein incorporated by reference).
  • II. Uniform Nanoemulsion Pharmacokinetics
  • In one embodiment, the present invention contemplates a nanoemulsion produced by a continuous turbulent flow at high pressure having improved pharmacokinetic properties when compared to conventional nanoparticulate compositions and/or nanoemulsions currently known in the art. It is known that nanoparticles deliver and/or release drugs (i.e., for example, norflaxin) and/or proteins (i.e., for example, serum albumin) more effectively than microparticles. Jeon et al., “Effect of solvent on the preparation of surfactant-free poly(DL-lactide-co-glycolide) nanoparticles and norfloxacin release characteristics’ Int J Pharm 207; 99-108 (2000); and Panyam et al., “Polymer degradation and in vitro release of a model protein from poly(D,L-lactide-co-glycolide) nano- and microparticles” J Control Release 92:173-187 (2003).
  • One embodiment of the present invention contemplates a uniform microfluidized nanoemulsion having improved pharmacokinetic properties when compared to conventional nanoparticulate compositions and/or nanoemulsions currently known in the art. One advantage of uniform microfluidized nanoemulsions comprises a narrow particle diameter range (i.e., for example, 10-110 nm). Most conventional nanoparticle compositions and/or nanoemulsions currently known have a wide distribution of particle diameters that interfere with the improved efficacies and bioavailabilities of the smaller sized particles.
  • The present invention has solved the problem of generating nanoemulsions with highly variable particle diameters and provides a more uniformly small-sized nanoemulsions (i.e., for example, a uniform nanoemulsion comprising stable particles). Consequently, these uniform nanoemulsions provide improved pharmacokinetic parameters when compared to conventional nanoparticle compositions and/or nanoemulsions currently known in the art independent of the mode of delivery which includes, but is not limited to, oral, transdermal, intravenous, intraperitoneal, intramuscular, subcutaneous, etc.
  • A. Absorption Phase
  • The use of conventional nanoparticulate compositions or nanoemulsions is not ideal due to delayed onset of action. In contrast, a uniform microfluidized nanoemulsion as contemplated by the present invention exhibits faster therapeutic effects.
  • Pharmaceuticals and nutraceuticals are commercially available as tablets, liquids, gel caps, capsules etc., generally intended for oral administration. Peak plasma concentrations of these compositions usually occur between 2-4 hours following administration.
  • When a uniform microfluidized nanoemulsion contemplated by the present invention is formulated into an oral dosage form peak plasma concentrations of an incorporated compound can be obtained in less than about 2 hours, preferably less than about 1 hour, more preferably less than about 30 minutes, but most preferably between 1 and 15 minutes.
  • B. Frequency of Dosing and Dosage Quantity
  • The recommended total daily dose of most pharmaceuticals and nutraceuticals are administered in divided doses. It is known in the art that a single daily dose may be preferable to multiple dose each day. For example, in studies of adults with partial onset seizures, a daily dose of 200 mg/day has inconsistent effects and is less effective than 400 mg/day. See Physicians' Desk Reference, 57.sup.th Edition, pp. 2502 (2003).
  • In contrast, some uniform microfluidized nanoemulsions of the present invention may be administered less frequently, at lower doses, and in dosage forms such as liquid dispersions, powders, sprays, solid re-dispersible dosage forms, ointments, creams, etc. Exemplary types of formulations useful in the present invention include, but are not limited to, liquid dispersions, gels, aerosols (pulmonary and nasal), ointments, creams, solid dose forms, etc. of any pharmaceutical, nutraceutical, or cosmeceutical. Lower dosages can be used because the smaller particle diameters of embodiments of the present invention ensure more complete absorption.
  • In one embodiment, the present invention contemplates a therapeutically effective amount of a uniform microfluidized nanoemulsion having ⅙, ⅕, ¼, ⅓, or ½ of the therapeutically effective amount of a conventional pharmaceutical, nutraceutical, or cosmeceutical formulations.
  • C. Oral Administration
  • A liquid dosage form of a conventional nanoparticulate or nanoemulsion composition would be expected to be a relatively large volume, highly viscous substance which would not be well accepted by subject populations. Moreover, viscous solutions can be problematic in parenteral administration because these solutions require a slow syringe push and can stick to tubing. In addition, conventional formulations of poorly water-soluble active agents tend to be unsafe for intravenous administration techniques, which are used primarily in conjunction with highly water-soluble substances. Embodiment contemplated by the present invention solves this problem by utilizing a liquid dispersion medium in which the pharmaceutical, nutraceutical, or cosmeceutical is substantially soluble.
  • Liquid dosage forms of embodiments of a uniform microfluidized nanoemulsion provide significant advantages over a liquid dosage form of a conventional nanoparticulate or nanoemulsion. In one embodiment, the uniform microfluidized nanoemulsion comprises a low viscosity. In another embodiment, the uniform nanoemulsion comprises a silky texture. These advantages include, for example: i) better subject compliance due to the perception of a lighter formulation which is easier to consume and digest; ii) ease of dispensing because one can use a cup or a syringe; iii) potential for formulating a higher concentration of a pharmaceutical, nutraceutical, or cosmeceutical resulting in a smaller dosage volume and thus less volume for the subject to consume; and iv) easier overall formulation concerns.
  • Liquid formulations of uniform nanoemulsions contemplated by the present invention are easier to consume which is especially important when considering juvenile subjects, terminally ill subjects, and elderly subjects. Viscous or gritty formulations, and those that require a relatively large dosage volume, are not well tolerated by these subject populations. Liquid oral dosage forms can be particularly preferably for subject populations who have difficulty consuming tablets, such as infants and the elderly.
  • The viscosities of liquid dosage forms of nanoparticulate topiramate according to the invention are preferably less than about 1/200, less than about 1/175, less than about 1/150, less than about 1/125, less than about 1/100, less than about 1/75, less than about fraction 1/50, or less than about 1/25 of a liquid oral dosage form of a conventional nanoparticulate composition or nanoemulsion at about the same concentration per ml.
  • In one embodiment, the present invention contemplates a uniform microfluidized nanoemulsion that is not turbid. In one embodiment, turbid refers to the property of particulate matter that can be seen with the naked eye or that which can be felt as “gritty” when consumed. Embodiments of nanoemulsions contemplated by the present invention can be poured out of or extracted from a container as easily as water, whereas a liquid dosage form of a conventional nanoparticulate or nanoemulsion composition is expected to exhibit notably more “sluggish” characteristics.
  • D. Increased Bioavailability
  • In one embodiment, the present invention contemplates a uniform microfluidized nanoemulsion having an increased bioavailability and a smaller dose requirement as compared to prior conventional nanoparticulate compositions and nanoemulsions administered at the same dose.
  • Any pharmaceutical, nutraceutical, or cosmeceutical can have adverse side effects if administered at a specific dose for a specific duration. Thus, lower doses which can achieve the same or better therapeutic effects as those observed with larger doses are desired. Such lower doses may be realized with a uniform microfluidized nanoemulsion contemplated by the present invention due to greater bioavailability as compared to conventional nanoparticulate compositions and nanoemulsions; consequently smaller dose of pharmaceuticals and nutraceutical are likely required to obtain the desired therapeutic effect.
  • For example, the relative bioavailability of pharmaceutical, nutraceutical, or cosmeceutical incorporated into a conventional nanoparticulate or nanoemulsion may be about 85% (i.e., as compared to a pure solution). In one embodiment, a uniform microfluidized nanoemulsion formulated into an oral pharmaceutical, nutraceutical, or cosmeceutical dosage form has a relative bioavailability preferably greater than about 85%. In other embodiments, the relative bioavailability is greater than about 90%, or greater than about 95%, or greater than about 98%.
  • E. Pharmacokinetic Profiles
  • The present invention also provides embodiments of uniform microfluidized nanoemulsions having incorporated pharmaceuticals and/or nutraceuticals having improved pharmacokinetic profiles when administered to mammalian subject. In one embodiment, the improved profile is compared to conventional nanoparticulate compositions and nanoemulsions.
  • An improved pharmacokinetic (pK) profile according to the present invention can have several different types of attributes. In one embodiment, an improved pK profile of a uniform microfluidized nanoemulsion may produce the same pK profile as a conventional nanoparticulate composition or nanoemulsion, but at a lower dose. In another embodiment, an improved pK profile requires less frequent dosing as compared to a conventional nanoparticulate composition or nanoemulsion. In one embodiment, an improved pK profile shows a faster onset of activity and/or greater quantity of drug absorbed (i.e., greater bioavailability) than conventional nanoparticulate compositions and nanoemulsions. In another embodiment, an improved pK profile allows a more effective and/or faster titration of the subject to therapeutic plasma levels.
  • The present invention contemplates certain embodiments of uniform microfluidized nanoemulsions comprising an improved pharmacokinetic profile as reflected by time-to-maximum-concentration (Tmax), maximum-concentration (Cmax), and/or area-under-curve (AUC) profiles.
  • In one embodiment, an administered dose of a pharmaceutical, nutraceutical, or cosmeceutical incorporated into a uniform microfluidized nanoemulsion comprises a Tmax less than that of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage. Preferably the Tmax is less than about 99%, less than about 90%, less than about 80%, less than about 70%, less than about 60%, less than about 50%, less than about 40%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, or less than about 10% of the Tmax of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • In another embodiment, an administered dose of a pharmaceutical, nutraceutical, or cosmeceutical incorporated into a uniform microfluidized nanoemulsion comprises a Cmax greater than that of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage. Preferably, the Cmax is greater than about 5%, greater than about 10%, greater than about 15%, greater than about 20%, greater than about 30%, greater than about 40%, greater than about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, greater than about 100%, greater than about 110%, greater than about 120%, greater than about 130%, greater than about 140%, or greater than about 150% than the Cmax of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • In one embodiment, an administered dose of a pharmaceutical, nutraceutical, or cosmeceutical incorporated into a uniform microfluidized nanoemulsion comprises an AUC greater than that of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage. Preferably, the AUC is greater than about 5%, greater than about 10%, greater than about 15%, greater than about 20%, greater than about 30%, greater than about 40%, greater than about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, greater than about 100%, greater than about 110%, greater than about 120%, greater than about 130%, greater than about 140%, or greater than about 150% than the AUC of a conventional nanoparticulate composition and/or nanoemulsion, administered at the same dosage.
  • III. Sterile Nanoemulsions
  • The present invention contemplates a method of making a nanoemulsion having anti-bacterial properties. In one embodiment, the method comprises a step exposing a premix to a continuous turbulent flow at high pressure. In one embodiment, the anti-bacterial nanoemulsion is prepared by microfluidization. In one embodiment, the exposing comprises approximately thirty (30) seconds. In another embodiment, the exposing comprises a pressure of at least 25,000 PSI. In another embodiment, the anti-bacterial nanoemulsion comprises soy protein.
  • For example, a powdered soy protein preparation was added to water thus creating a suspension. Then, a first aliquot of the suspension was added to a first container (i.e., for example, a cell culture falcon flask) that served as a control. A second aliquot of the suspension was microfluidized (supra) to create a nanoemulsion. The preparation was microfluidized in accordance with Example 5. The microfluidized nanoemulsion was then added to a second container. Both containers were refrigerated immediately and observed over the next several days. The control suspensions agglomerated and grew bacteria. See FIGS. 15A and 15B. In contrast, the microfluidized nanoemulsion containing the soy protein did not agglomerate or grow bacteria. See FIGS. 15C and 15D.
  • Although it is not necessary to understand the mechanism of an invention, it is believed that the microfluidization sterilized the bacteria. It is further believed that the microfluidization shear stress resulted in a bacterial cell lysis thereby preventing further bacterial growth. Consequently, it is believed that microfluidization, as contemplated herein, produces a microbiologically sterile composition.
  • In one embodiment, the present invention contemplates a nanoemulsion comprising an oxidizing environment produced by a method comprising a continuous turbulent flow at a high pressure. In one embodiment, the nanoemulsion comprises a uniform microfluidized nanoemulsion. In one embodiment, the oxidizing environment prevents bacterial growth. In another embodiment, the oxidizing environment is bacteriocidal. In another embodiment, the oxidizing environment provides a sterile nanoemulsion.
  • An oxidizing nanoemulsion environment may result from an increased surface to volume ratio. In one embodiment, the present invention also contemplates a method to avoid the generation of an oxidizing environment by microfluidizing in the presence of an antioxidant. In one embodiment, the antioxidant reduces the presence of reactive oxygen species (ROS) in the microfluidized nanoemulsion. In another embodiment, the
  • Sample Sample Sample
    Formulation 1 2 3 Mean
    Plasma 4.0 2.8 3.3 3.4
    (unoxidized control)
    Plasma + FeCl3 12.4 16.0 13.1 13.9
    (oxidized control)
    1.5 g DHA with 200 ml milk 44.0 42.6 45.8 44.1
    (microfluidized)
    1.75 g DHA, 1000 mg Vit E 12.8 19.2 20.1 17.4
    and 800 mg Vit C with 200 ml milk
    (not microfluidized)
    1.75 g DHA, 1000 mg Vit E 4.0 6.0 3.5 4.5
    and 800 mg Vit C with 200 ml milk
    (microfluidized)
    1.75 g DHA and 800 mg Vit C 17.5 17.8 20.8 18.7
    with 200 ml milk (microfluidized)
    1.75 g DHA and 1000 mg Vit E 9.8 16.7 11.4 12.6
    with 200 ml milk (microfluidized)

    antioxidants are encapsulated by the nanoparticles for subsequent release to the subject.
  • The ROS load within any nanoemulsion preparation can be quantitatively determined by measuring indicators of an oxidizing environment. Malondialdehyde (MDA), is a known indicator of an oxidizing environment.
  • Table 1: Oxidative Stress in Nanoemulsion Formulations as Measured by Malondialdehyde Formation
  • As can be seen in Table 1 above, the process of making a microfluidized nanoemulsion increases MDA levels by approximately 13-fold. Further, the presence of both vitamin C and/or vitamin E completely prevented MDA generation in microfluidized nanoemulsions thereby returning MDA to homeostatic plasma levels.
  • EXPERIMENTAL
  • The following examples are specific embodiments as contemplated by the present invention and are not intended to be limiting.
  • Example 1 Stable Formulation of Plant Sterol Microfluidized Nanoemulsions
  • This example presents one plant sterol embodiment of a microfluidized nanoemulsion. The step-wise procedure is as follows:
  • 1. Heat 4 g of soybean oil
  • 2. Add 5 g soy lecithin, stir and heat to 90° C.
  • 3. Add 1 g plant sterol, stir and heat 10 mins
  • 4. Add 250 mg polysorbate 80.
  • 5. Heat 240 mL de-ionized water to 70° C.
  • 6. Add step 4 mixture to step 5 mixture, keep stir bar and heat on for 30 mins
  • 7. Homogenize step 6 mixture for 2-4 mins
  • 8. Stir formulation for 10 mins on hot plate
  • 9. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 10. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter (i.e., Peak 1/Peak 2) for these microfluidized plant sterol nanoemulsions was 39 nm. See FIG. 1. The average particle diameter data for the plant sterol microfluidized nanoemulsion is shown in Table 2 below.
  • TABLE 2
    Microfluidized Plant Sterol Nanoemulsion
    Diam. (nm) % Intensity Width (nm)
    Peak 1: 54.16 85.86 14.36
    Peak 2: 15.55 14.14 2.521
    Peak 3: 0 0 0
    Z-Average: 38.91;
    PDI: 0.228;
    Intercept: 0.9764.
  • After three months the particle diameter was again determined. The mean particle diameter (i.e., Peak 1) for this microfluidized plant sterol nanoemulsion was 64.4 nm. See FIG. 1A. The average particle diameter data for the three month plant sterol nanoemulsion is shown in Table 3 below.
  • TABLE 3
    Three Month Storage: Microfluidized Plant Sterol Nanoemulsion
    Diam. (nm) % Intensity Width (nm)
    Peak 1: 74.8 100 120.8
    Peak 2: 0 0 0
    Peak 3: 0 0 0
    Z-Average: 64.4;
    PDI: 0.196;
    Intercept: 0.969.
  • Example 2 Stable Formulation of Cod Liver Oil Microfluidized Nanoemulsions
  • This example presents one cod liver oil embodiment of a microfluidized nanoemulsion that has a stable particle diameter for at least four months. The step-wise procedure is as follows:
  • 1. Heat 5 g of soybean oil (65° C.)
  • 2. Add 5 g cod liver oil, stir and heat to 80° C.
  • 3. Add 6 g polysorbate 80, stir and heat 20 mins
  • 4. Add 200 mL de-ionized water, stir and heat 30 mins
  • 5. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 6. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter (i.e., Peak 1/Peak 2) for this cod liver oil microfluidized nanoemulsion was 58 nm. Before microfluidization, the mean particle diameter of the cod liver oil suspension was 2,842 nm. This represents a 50-fold reduction with a single pass through the microfluidizer. Four months after the microfluidization process, the particle diameter was again determined and found not to have changed. See FIG. 2. The average particle diameter data from the four-month microfluidized sample is presented in Table 4.
  • TABLE 4
    Microfluidized Cod Liver Oil Nanoemulsion Four Months
    After Preparation
    Diam. (nm) % Intensity Width(nm)
    Peak 1: 63.92 82.22 15.62
    Peak 2: 18.51 17.78 2.771
    Peak 3: 0 0 0
    Z-Average: 45.15;
    PDI: 0.247;
    Intercept: 0.9707.
  • Example 3 Stable Formulation of Tocopherol Microfluidized Nanoemulsions
  • This example presents one tocopherol embodiment of a microfluidized nanoemulsion that maintains particle diameter for at least five months. The step-wise procedure is as follows:
  • 1. Heat 13.5 g of soybean oil
  • 2. Add 2 g tocopherol, stir and heat to 90° C.
  • 3. Heat 2 g polysorbate 80 in 100 mL de-ionized water, heat to 75° C.
  • 4. Add step 3 mixture to step 2 mixture
  • 5. Heat 300 mL di-ionized water and 6 g polysorbate 80, heat till 70° C.
  • 6. Add step 4 mixture to step 5 mixture, keep stir bar and heat on
  • 7. Homogenize step 6 mixture for 2-4 mins
  • 8. Stir formulation for 3-5 mins on hot plate
  • 9. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 10. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter for the tocopherol microfluidized nanoemulsion was 64 nm. Before microfluidization, the mean particle diameter for the tocopherol suspension was 1,362 nm. This represents a 21-fold reduction a single pass through the microfluidizer. Five months after the microfluidization process, the particle diameter was again determined and found not to have changed. See FIG. 3. The average particle diameter data from the five-month microfluidized sample is presented in Table 5.
  • TABLE 5
    Microfluidized Tocopherol Nanoemulsion Five Months
    After Preparation
    Diam. (nm) % Intensity Width (nm)
    Peak 1 88.06 77.84 19.99
    Peak 2 26.46 22.16 3.651
    Peak 3 0 0 0
    Z-Average: 58.07;
    PDI: 0.234;
    Intercept: 0.9697
  • Example 4 Formulation of Lutein and Zeaxanthin Microfluidized Nanoemulsions
  • This example presents one lutein/zeaxanthin embodiment of a microfluidized nanoemulsion. The step-wise procedure is as follows:
  • 1. Heat 5 g of soybean oil
  • 2. Add 2 g of lecithin
  • 3. Heat and stir, 10 mins
  • 4. Add 125 mg of lutein and 125 mg of zeaxanthin
  • 5. Heat and stir, 10 mins
  • 6. Heat 240 ml de-ionized water, 50° C.
  • 7. Add heated water to mixture
  • 8. Stir and heat, till it is a solution
  • 9. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 10. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter (i.e., Peak 1/Peak 2) for the lutein and zeaxanthin microfluidized nanoemulsion was 62 nm. See FIG. 4. The average particle diameter data for the sample is shown in Table 6.
  • TABLE 6
    Microfluidized Lutein/Zeaxanthin Nanoemulsion
    Diam. (nm) % Intensity Width (nm)
    Peak 1 89.45 83.96 21.1
    Peak 2 22.81 16.04 2.968
    Peak 3 0 0 0
    Z-Average: 62.26
    PDI: 0.245
    Intercept: 0.976
  • Example 5 Formulation of Soy Protein Microfluidized Nanoemulsion
  • This example presents one soy protein embodiment of a microfluidized nanoemulsion. The step-wise procedure is as follows:
  • 1. Heat 5 g soybean oil
  • 2. Add 5 g liquid lecithin
  • 3. Heat and stir 10 mins, 70° C.
  • 4. Heat 240 mL de-ionized water, 65° C.
  • 5. Add heated water to mixture
  • 6. Add 9 g soy protein, stir and heat 10 min
  • 7. Add 9 g soy protein
  • 8. Stir and heat 20 min, 70° C.
  • 9. Homogenize 1 min
  • 10. Microfluidize using a M-110EH unit ten times at 25,000 PSI
  • 11. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter (i.e., Peak 1/Peak 2) for the vanilla soy protein (Central Soya) microfluidized nanoemulsion was 55 nm. See FIG. 5. The average particle diameter data for the sample is shown in Table 7.
  • TABLE 7
    Microfluidized Soy Protein Nanoemulsion
    Diam. (nm) % Intensity Width (nm)
    Peak 1 55.15 80.32 16.45
    Peak 2 290.8 19.68 82.82
    Peak 3 0 0 0
    Z-Average: 54.97;
    PDI: 0.283;
    Intercept: 0.9819
  • Example 6 Formulation of Whey Protein Microfluidized Nanoemulsion
  • This example presents one whey protein embodiment of a microfluidized nanoemulsion. The step-wise procedure is as follows:
  • 1, Heat 5 g soybean oil
  • 2. Add 5 g soy lecithin
  • 3. Add 250 mg polysorbate 80
  • 4. Heat and stir 10 mins, 70° C.
  • 5. Heat 240 mL de-ionized water, 65° C.
  • 6. Add heated water to mixture
  • 7. Add 10 g whey protein
  • 8. Stir and heat 10 min
  • 9. Homogenize 1 min
  • 10. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 11. Do particle diameter analysis Malvern Nano S instrument
  • The mean particle diameter (i.e., Peak 1/Peak 2) for the whey protein microfluidized nanoemulsion was 108 nm. See FIG. 6. The average particle diameter data for the sample is shown in Table 8.
  • TABLE 8
    Microfluidized Whey Protein Nanoemulsion
    Diam. (nm) % Intensity Width (nm)
    Peak 1 127.7 91.3 38.09
    Peak 2 23.72 6.161 2.764
    Peak 3 5027 2.536 593
    Z-Average: 108.2
    PDI: 0.263
    Intercept: 0.948
  • Example 7 Formulation of Orange Juice, Plant Sterol and Lutein Microfluidized Nanoemulsion
  • This example presents one orange juice/plant sterol/lutein embodiment of a microfluidized nanoemulsion. The step-wise procedure is as follows:
  • 1. Heat soybean oil, 80° C.
  • 2. Add 1.5 g plant sterol
  • 3. Stir and heat, 5 min
  • 4. Add 5 g polysorbate 80
  • 5. Add 70 mg Lutein
  • 6. Stir and heat, 10 min
  • 7. Add 240 mL orange juice (Tropicana®)
  • 8. Stir and heat, 1 hour
  • 9. Microfluidize using a M-110EH unit twice at 25,000 PSI
  • 10. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter (i.e., Peak 1/Peak 2) for the orange juice/plant sterol/lutein microfluidized nanoemulsion was 46 nm. See FIG. 7. The average particle diameter data for the sample is shown in Table 9.
  • TABLE 9
    Microfluidized Orange Juice/Plant Sterol/Lutein Nanoemulsion
    Diam. (nm) % Intensity Width (nm)
    Peak 1 61.55 81.57 15.32
    Peak 2 17.13 16.1 2.433
    Peak 3 5143 2.329 509.4
    Z-Average: 46.41;
    PDI: 0.322;
    Intercept: 0.9609
  • Example 8 Stable Formulation of DHA Fish Oil/Water Microfluidized Nanoemulsion
  • This example presents one DHA fish oil/water embodiment of a microfluidized nanoemulsions that maintains particle diameter for at least two months. The step-wise procedure is as follows:
  • 1. Heat 6.4 g DHA fish oil
  • 2. Add 6 g soy lecithin
  • 3. Add 250 mg polysorbate 80
  • 4. Heat 240 mL de-ionized water, 75° C.
  • 5. Add heated water to mixture
  • 6. Stir and heat, 20 mins
  • 7. Homogenize 2 mins
  • 8. Stir and heat, 10 mins
  • 9. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 10. Do particle diameter analysis using a Malvern Nano S instrument.
  • The mean particle diameter (i.e., Peak 1) for the DHA fish oil/water microfluidized nanoemulsion was 73 nm. Two months after the microfluidization process, the particle diameter was again determined and found not to have changed. See FIG. 8. The average particle diameter data from the two-month microfluidized sample is presented in Table 10.
  • TABLE 10
    Stable Microfluidized DHA Fish Oil/Water Nanoemulsion
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 81.73 100 20.38
    Peak 2 0 0 0
    Peak 3 0 0 0
    Z-Average: 72.58;
    PDI: 0.205;
    Intercept: 0.9636.
  • Example 9 Stable Formulation of DHA Fish Oil/Milk Microfluidized Nanoemulsion
  • This example presents one DNA fish oil/milk embodiment without any added emulsifiers that maintains particle diameter for at least three (3) weeks. The step-wise procedure is as follows:
  • 1. Heat 1.5 g DHA fish oil, 50° C.
  • 2. Heat 200 mL whole milk, 50° C.
  • 3. Mix the two together
  • 4. Stir and heat, 10 mins
  • 5. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 6. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter (i.e., Peak 1) for the DHA fish oil/milk microfluidized nanoemulsion 93 nm. This nano-emulsion preparation was made without any added emulsifiers. Three weeks after the microfluidization process, the fish oil was still in solution and the particle diameter was again determined and found not to have changed. See FIG. 9. The average particle diameter data from the three-week microfluidized sample is presented in Table 11.
  • TABLE 11
    Stable Microfluidized DNA Fish Oil/Milk Nanoemulsion
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 106.9 100 32.84
    Peak 2 0 0 0
    Peak 3 0 0 0
    Z-Average: 93.11;
    PDI: 0.178;
    Intercept: 0.9341
  • Example 10 Temperature Stability of Microfluidized Nanoemulsions
  • This example presents the stability of microfluidized nanoemulsions following exposure to either heat or cold. The formulation used in this experiment comprised DHA Fish Oil milk/tocopherol.
  • 1. Dissolved 1 g of vitamin C in 25 mL of di-ionized water
  • 2. Added 200 mL of whole milk to step 1
  • 3. Took 1.7 g DHA fish oil and added 800 mg of delta tocopherol
  • 4. Added steps 1 and 2 to step 3
  • 5. Stir and heat 10 mins, 50° C.
  • 6. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 7. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter (i.e., Peak 1) for the DHA fish oil/milk/tocopherol microfluidized nanoemulsion was 87 nm. See FIG. 10. This nano-emulsion preparation was made without any added emulsifiers. The average particle diameter data for the original nanoemulsion is presented in Table 12.
  • TABLE 12
    Microfluidized DNA Fish Oil/Milk/Tocopherol Original Nanoemulsion
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 91.84 97.86 23.95
    Peak 2 5179 2.144 485.1
    Peak 3 0 0 0
    Z-Average: 87.09;
    PDI: 0.216;
    Intercept: 0.9339
  • This original microfluidized nanoemulsion was pasteurized at 75° C. for 30 seconds. Twenty-four hours later, the oil was still in solution and the particle diameter was stable as compared to the original nanoemulsion. See FIG. 11. The average particle diameter data for the pasteurized microfluidized nanoemulsion is presented in Table 13.
  • TABLE 13
    Microfluidized DHA Fish Oil/Milk/Tocopherol Pasteurized Nanoemulsion
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 108.3 82.49 28.06
    Peak 2 45.16 17.51 8.109
    Peak 3 0 0 0
    Z-Average: 87.18;
    PDI: 0.198;
    Intercept: 0.9281
  • The original microfluidized nanoemulsion was freeze-thaw tested at −4° C. for 24 hours. Twenty-four hours later, the oil was still in solution and the particle diameter was stable as compared to the original nanoemulsion. See FIG. 12. The average particle diameter data for the freeze-thaw microfluidized nanoemulsion is presented in Table 14.
  • TABLE 14
    Microfluidized DHA Fish Oil/Milk/Tocopherol Freeze-Thaw
    Nanoemulsion
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 99.72 100 39.07
    Peak 2 0 0 0
    Z-Average: 87.58;
    PDI: 0.198
  • Example 11 Improved Bioavailability of Dietary Lycopene
  • This example demonstrates an improved bioavailability of lycopene when fed as a uniform microfluidized nanoemulsion versus mixed into a standard diet formulation.
  • The lycopene microfluidized nanoemulsion was prepared in a step-wise manner as follows:
  • 1. Heat 5 g of soybean oil
  • 2. Add 2 g of lecithin
  • 3. Heat and stir, 10 mins
  • 4. Add 125 mg of lycopene
  • 5. Heat and stir, 10 mins
  • 6. Heat 240 ml de-ionized water (or grape juice); 50° C.
  • 7. Add heated water (or grape juice) to mixture
  • 8. Stir and heat, till it is a solution
  • 9. Microfluidize using a M-110EH unit once at 25,000 PSI
  • 10. Do particle diameter analysis using a Malvern Nano S instrument
  • The mean particle diameter for the lycopene microfluidized nanoemulsion was 74 nm.
  • Bioavailability In Gerbils
  • The microfluidized nanoemulsion was incorporated into a chow-based diet and fed to gerbils over a 4 week period. A control group was fed a lycopene in oil-enriched chow-based diet. At the end of 4 weeks, blood was collected, plasma harvested and plasma lycopene levels were determined by HPLC in both gerbil groups.
  • FIG. 13 demonstrates that control gerbils did not demonstrate detectable plasma lycopene levels. The gerbils fed a chow comprising a microfluidized lycopene nanoemulsion, however, demonstrated elevated plasma lycopene levels. See FIG. 14,
  • Bioavailability In Humans
  • A microfluidized lycopene nanoemulsion was then prepared with grape juice instead of water and orally administered to two (2) human subjects over a 4 day period (125 mg/serving, 2 servings per day). This administration raised plasma lycopene levels by approximately 38% (data not shown).
  • Example 12 Improved Efficacy of Microfluidized Nanoemulsions
  • This example presenting data showing that microfluidized nanoemulsions provide improved efficacy over that seen in traditional nanoemulsions. Specifically, this example compares the ability of three plant sterol formulations to reduce plasma low density lipoprotein cholesterol (LDL-C) levels in hypercholesterolemic hamsters.
  • A microfluidized mixed plant sterol (60% sitosterol) nanoemulsion was prepared in a step-wise manner as follows:
  • 1. Heat 5 g soybean oil.
  • 2. Add 5 g soy lecithin, stir and heat 15 mins.
  • 3. Repeat Step 2.
  • 4. Add 15 g soybean oil, stir and heat 10 mins.
  • 5. Add 4 g plant sterol, stir and heat 10 mins.
  • 6. Repeat Step 4 four (4) times.
  • 7. Add 1 g polysorbate 80, stir and heat 10 mins.
  • 8. Heat 200 ml MinuteMaid Heartwise® orange juice (75° C.).
  • 10. Heat 1800 ml MinuteMaid Heartwise® orange juice (70° C.).
  • 11. Add Step 8 to Step 7. Stir and heat 20 min (80° C.).
  • 12. Add to Step 10.
  • 13. Add 1 g polysorbate 80, stir and heat 20 min (80° C.).
  • 14. Homogenize for 2-4 min.
  • 15. Stir homogenate on hot plate for 10 min.
  • 16. Microfluidize using a M-110EH unit at 25,000 PSI.
  • 17. Do particle analysis using a Malvern Nano S instrument.
  • The mean particle diameter for the microfluidized plant sterol nanoemulsion was 41.95 nm. See FIG. 16.
  • Forty (40) hamsters were divided into four (4) groups of ten (10) each. Group I was fed a control hypercholesterolemic diet (HCD); Group 2 was fed 30 mg/d of crystalline plant sterol; Group 3 was fed 20 mg/d of MinuteMaid Heartwise® micronized plant sterol (Cargill); Group 4 was fed 10 mg/d of the microfluidized plant sterol nanoemulsion. After four (4) weeks, blood samples were analyzed for plasma LDL-C levels. The microfluidized plant sterol nanoemulsion was twice as effective as the MinuteMaid Heartwise® micronized diet, and three times as effective as the crystalline plant sterol diet. See FIG. 17.
  • The data show that the improved bioavailability shown in Example 11 results in improved clinical therapy when compared to micron-sized or crystalline plant sterol diets.
  • Example 13 Cholesterol Nanoemulsions Insoluble vs Soluble Dispersion Media
  • This example presents data demonstrating that uniform microfluidized nanoemulsion compositions depend upon a compound having substantial solubility in the liquid dispersion medium. This example compares the microfluidizing technique described in U.S. Pat. No. 5,510,118 to one embodiment as contemplated by the present invention. The absorbable lipid cholesterol was chosen as the test compound.
  • Group I represents the '118 premix and was prepared by dispersing cholesterol (2 gms), water (100 mls) and Tween® 80 (0.2 gms), where cholesterol is insoluble (i.e., below at least 30 mg/ml) in the liquid dispersion medium (water). Thereafter, this cholesterol/water/Tween® 80 solution was microfluidized using a M-100EH unit. Multiple passes (10-15) through the microfluidizer were performed at PSI's ranging between 4,000-20,000 but were terminated because the generated heat exceeded 70° C. (much higher than the recommended 30-40° C. in the '118 patent. After the microfluidization it was observed that much of the cholesterol had precipitated. After twenty-four hours, the preparation of the Group I nanoemulsion contained only 0.44 gms (i.e., 22%) of the original cholesterol weight.
  • Group II represents one embodiment of the present invention and was prepared by dispersing cholesterol (2 gms) in heated soybean oil (10 gms), soy lecithin (5 gms), and Tween® 80 (0.2 gms) where cholesterol is substantially soluble (i.e., above at least 30 mg/ml) in the dispersion medium (oil). Thereafter, this cholesterol/oil/lecithin/Tween® 80 was added to 100 ml of heated water and microfluidized using a single 30 second pass at 25,000 PSI using a M-100EH unit. After the microfluidization cholesterol precipitation was not noticeably evident. After twenty-four hours, the preparation of the Group II nanoemulsion contained 1.66 gm (i.e., 83%) of the original cholesterol weight.
  • The data show that the particle diameter distributions from both Group I and Group II premix preparations are practically identical. See FIG. 18A and FIG. 18B, respectively. Specifically, a single peak ranging from 700-1000 nm having a mean particle diameter of approximately 900 nm is observed for both preparations. See Tables 15 and 16.
  • TABLE 15
    Cholesterol/Tween ® 80/Water Premix Particle diameter: Group I
    Diam. (nm) % Intensity Width (nm)
    Peak 1 942.5 100 38.9
    Z-Average: 1982;
    PDI: 0.210;
    Intercept: 0.6797
  • TABLE 16
    Cholesterol/Oil/Lecithin/Tween ® 80/Water Premix Particle
    diameter: Group II
    Diam. (nm) % Intensity Width (nm)
    Peak 1 897.9 100 64.8
    Z-Average: 1328;
    PDI: 0.427;
    Intercept: 0.6989
  • Following microfluidization, however, the particle diameter distributions are vastly different between Group I and Group II. See FIG. 19A and FIG. 19B, respectively. Group I shows two vastly disparate and distinct peaks. See Table 17. Group II, however, shows a single peak representing one embodiment of a uniform microfluidized nanoemulsion. See Table 18.
  • TABLE 17
    Microfluidized Cholesterol/Tween ® 80/Water Nanoemulsion
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 578.2 67.6 120.8
    Peak 2 96.7 32.3 14.9
    Z-Average: 246.5;
    PDI: 0.789;
    Intercept: 0.7687
  • TABLE 18
    Microfluidized Cholesterol/Oil/Lecithin/Tween ® 80/Water
    Nanoemulsion
    Diam. (nm) % Intensity Width (nm)
    Peak 1 101.3 100 25.1
    Z-Average: 86.8;
    PDI: 0.240;
    Intercept: 0.9455
  • The data above demonstrate that some embodiments of the present invention contemplate improvements over the art in creating uniform microfluidized nanoemulsions. In particular, it is now clear that the Bosch et al ('118 patent), and the Cooper et al. portfolio ('758, '038, and '202 application publications) do not teach a microfluidization process that creates a uniform particle diameter distribution.
  • Example 14 Nanoparticulate Compositions vs Uniform Microfluidized Nanoemulsions
  • This example describes a demonstration that will show that a milled nanoparticle composition (for example, one made according to US Appln Publ No. 2004/0033202 to Cooper et al.) does not create a uniform particle diameter distribution as does a microfluidized nanoemulsion as contemplated by one embodiment of the present invention. An absorbable phytosterol will be chosen as the test compound.
  • Group I represents the '202 premix that will be prepared by dispersing 5% (w/w) phytosterol/water solution with 1% (w/w) Tween® 80, where the phytocholesterol is insoluble (i.e., below at least 30 mg/ml) in the liquid dispersion medium (water). Thereafter, this phytosterol/water/Tween® 80 solution will be milled at 10° C. for 1.5 to 2 hours in a DYNO®-Mill KDL (Willy A Bachofen A G, Machinefabrik, Basel, Switzerland) using a 500 μm milling media (i.e., grinding beads) of type Polymill® 500. After the milling it will be observed that much of the phytocholesterol has precipitated. After at least twenty-four hours, the preparation of the Group I nanoparticulate will contain less than ½ of the original phytosterol weight.
  • Group II represents one embodiment of the present invention and will be prepared by dispersing 5% (w/w) phytosterol/heated soybean oil solution, soy lecithin, with 1% Tween® 80, where the phytosterol is substantially soluble (i.e., above at least 30 mg/ml) in the liquid dispersion medium (oil). Thereafter, this phytosterol/oil/lecithin/Tween® 80 premix is added to 100 ml heated water and microfluidized using a single 30 second pass at 25,000 PSI using a M-100EH unit. After the microfluidization phytosterol precipitation will not be noticeably evident. After twenty-four hours, the preparation of the Group II nanoemulsion will contain greater than ¾ of the original phytosterol weight.
  • The data will show that the particle diameter distributions from both Group I and Group II premix preparations are practically identical. For example, a single peak ranging from 700-1000 nm having a mean particle diameter of approximately 900 nm might be observed for both preparations. See Tables 19 and 20.
  • TABLE 19
    Phytosterol/Tween 80/Water Premix Particle diameter: Group I
    Diam. (nm) % Intensity Width (nm)
    Peak 1 942.5 100 38.9
    Z-Average: 1982;
    PDI: 0.210;
    Intercept: 0.6797
  • TABLE 20
    Phytosterol/Oil/Lecithin/Tween 80/Water Premix Particle
    diameter: Group II
    Diam. (nm) % Intensity Width (nm)
    Peak 1 897.9 100 64.8
    Z-Average: 1328;
    PDI: 0.427;
    Intercept: 0.6989
  • Following processing however, the particle diameter distributions are expected to be vastly different between Group I and Group II. For example, Group I will most likely show at least two vastly disparate and distinct peaks. See Table 21. Group II, however, will have only a single peak representing one embodiment of a uniform microfluidized nanoemulsion. See Table 22.
  • TABLE 21
    Microfluidized Cholesterol/Tween 80/Water Nanoemulsion: Group I
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 578.2 67.6 120.8
    Peak 2 96.7 32.3 14.9
    Z-Average: 246.5;
    PDI: 0.789;
    Intercept: 0.7687
  • TABLE 22
    Microfluidized Cholesterol/Oil/Lecithin/Tween 80/Water
    Nanoemulsion: Group II
    Diam. (nm) % Intensity Width (nm)
    Peak 1 101.3 100 25.1
    Z-Average: 86.8;
    PDI: 0.240;
    Intercept: 0.9455
  • The data above demonstrate that nanoparticulate composition are not able to create uniform particle diameter distributions as contemplated by some embodiments of the nanoemulsions contemplated herein. In particular, it is now clear that the Cooper et al. portfolio ('758, '038, and '202 application publications) do not teach a milling process that creates a uniform particle diameter distribution.
  • Example 15 Improved Bioavailability Over Conventional Nanoparticulate Compositions
  • This example will provide data showing that a uniform microfluidized nanoemulsion as contemplated by one embodiment of the present invention has improved plant sterol bioavailability and/or efficacy than a conventional nanoparticulate composition.
  • A standard curve will be constructed by gavaging thirty (30) hamsters with 1 μCi 3H-cholesterol. Plasma cholesterol levels are then determined at Day 1, Day 2, Day 4, and Day 7. These data are used to calculate bioavailability of 3H-cholesterol during the 7 Day period as area-under-the-curve (AUC).
  • After plasma radioactivity levels have returned to background levels (i.e., approximately 7.5 cholesterol metabolic half-lives), the experiment will be repeated using the following treatment groups (n=10).
      • Group I: Standard diet mixed with a plant sterol.
      • Group II: Standard diet mixed with a uniform microfluidized plant sterol nanoemulsion prepared in accordance with Example 1.
      • Group III: Standard diet mixed with a conventional lycopene nanoparticulate composition prepared in accordance with conventional milling grinder techniques as described in the '202 Cooper et al. application.
  • The AUC measurement will determine the ability of each preparation to reduce the absorption of 3H-cholesterol into the bloodstream which is proportional to the bioavailability and/or efficacy of each preparation.
  • A greater bioavailability and/or efficacy of a plant sterol when administered as a uniform microfluidized nanoemulsion will be seen because: i) the average particle diameter of the uniform microfluidized nanoemulsion is smaller than the conventional nanoparticulate composition (i.e., for example, 300 nm v. 50 nm); ii) microfluidization produces more stable particles than either milling or homogenization; and iii) microfluidization produces pH-resistant particles (i.e., stomach acid or small intestine base conditions) unlike those produced by either milling or homogenization.
  • Example 16 Improved Efficacy Over Conventional Nanoparticulate Compositions
  • This example will provide data showing that a uniform microfluidized nanoemulsion as contemplated by one embodiment of the present invention has improved efficacy in lowering plasma cholesterol levels that a conventional nanoparticulate composition.
  • The study will have duration of six (6) weeks. Briefly, seventy (70) hamsters will be fed a liquid-based hypercholesterolemic diet for a two (2) week pre-test period in order to elevate and stabilize plasma cholesterol levels. Subsequently, the hamsters are divided into the seven (7) test groups (n=10) shown below. Each group is maintained on the liquid-based hypercholesterolemic diet and: i) a nanoparticulate composition (i.e., for example, prepared as per the '202 Cooper et al. application); or ii) a uniform microfluidized nanoemulsion as contemplated by one embodiment of the present invention, for four (4) additional weeks.
      • Group 1: Hypercholesterolemic diet only
      • Group II: Hypercholesterolemic diet+0.1% (w/w) plant sterol nanoparticulate composition.
      • Group III: Hypercholesterolemic diet+0.5% (w/w) plant sterol nanoparticulate composition.
      • Group IV: Hypercholesterolemic diet+1% (w/w) plant sterol nanoparticulate composition.
      • Group V: Hypercholesterolemic diet+0.1% (w/w) plant sterol uniform microfluidized nanoemulsion.
      • Group VI: Hypercholesterolemic diet+0.5% (w/w) plant sterol uniform microfluidized nanoemulsion.
      • Group VII: Hypercholesterolemic diet+1% (w/w) plant sterol uniform microfluidized nanoemulsion.
  • Blood samples are taken at 0, 2, 3, 4, 5, and 6 weeks where plasma cholesterol levels will be determined by methods known in the art.
  • A greater efficacy of the plant sterol uniform microfluidized nanoemulsions to lower plasma cholesterol levels is seen because: i) the average particle diameter of the uniform microfluidized nanoemulsion is smaller than the conventional nanoparticulate composition (i.e., for example, 300 nm v. 50 nm); ii) microfluidization produces more stable particles than either milling or homogenization; and iii) microfluidization produces pH-resistant particles (i.e., stomach acid or small intestine base conditions) unlike those produced by either milling or homogenization.
  • Example 17 Microfluidization Single Pass Comparison
  • This example provides data showing that the Bosch technique does not produce a uniform microfluidized nanoemulsion when compared to one embodiment of the present invention under identical microfluidization techniques.
  • The Group I & II premixes were prepared in accordance with Example 13. Each premix was subjected to one pass at 25,000 PSI in the microfluidizer. Group I (representing the Bosch formulation) shows that 85% of the particles have a mean diameter of 815 nm. See FIG. 20A. Group II (representing one embodiment of the present invention) shows that 98% of the particles have a mean diameter of 78 nm, See FIG. 20B This represents a greater than ten-fold difference in average diameter. Significantly, only 15% of the Bosch particles are within the 100 nm range, thereby representing a six-fold difference in particle diameter distribution in this lower range.
  • The average particle diameter distributions between Group I and Group II are presented in Tables 23 & 24 below.
  • TABLE 23
    Microfluidized Cholesterol/Tween ® 80/Water
    Nanoemulsion: Single Pass
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 815.3 84.5 117.7
    Peak 2 101.8 15.54 10.54
    Z-Average: 651.5;
    PDI: 84.5;
    Intercept: 0.7487
  • TABLE 24
    Microfluidized Cholesterol/Oil/Lecithin/Tween ® 80/Water
    Nanoemulsion: Single Pass
    Diam.
    (nm) % Intensity Width (nm)
    Peak 1 78.43 97.47 31.43
    Peak 2 19.63 2.535 2.928
    Z-Average: 65.98;
    PDI: 0.190;
    Intercept: 0.9210

Claims (31)

1. A nanoemulsion comprising a population of particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers.
2. The nanoemulsion of claim 1, wherein said particles encapsulate a compound.
3. The nanoemulsion of claim 2, wherein said compound is a pharmaceutical.
4. The nanoemulsion of claim 2, wherein said compound is a nutraceutical.
5. A nanoemulsion comprising a first and second population of particles, wherein the majority of particles in said first population have diameters between approximately 10 and approximately 20 nanometers, wherein the majority of particles in said second population have diameters between approximately 40 and approximately 80 nanometers, wherein said nanoemulsion is uncontaminated by particles having diameters larger than 110 nanometers.
6. The nanoemulsion of claim 5, wherein said particles encapsulate a compound.
7. The nanoemulsion of claim 6, wherein said compound is a pharmaceutical.
8. The nanoemulsion of claim 6, wherein said compound is a nutraceutical.
9. A method, comprising:
a) providing;
i) a premix comprising a compound and a liquid dispersion medium, wherein said compound has a solubility greater than 30 mg/ml in said medium; and
ii) a microfluidizer capable of maintaining at least 25,000 PSI;
b) using a single pass exposure of said premix to said microfluidizer to create a population of nanoemulsion particles having diameters ranging approximately between 10-110 nm.
10. The method of claim 9, wherein said dispersion medium is selected from the group consisting of aqueous media and oil-based media.
11. The method of claim 10, wherein said aqueous media is selected from the group consisting of water, saline solution, ringers solution, dextrose, and short chain alcohols.
12. The method of claim 10, wherein said oil-based media is selected from the group consisting of saturated and unsaturated oils from vegetable and marine sources, silicone oils, and mineral oils.
13. The method of claim 9, wherein said compound is selected from the group consisting of a plant sterol, cod liver oil, tocopherol, lecithin, lutein, zeaxanthin, and soy protein.
14. A method, comprising:
a) providing;
i) a premix comprising a compound, a first antioxidant, a second antioxidant, and an aqueous dispersion medium, wherein said compound has a solubility greater than 30 mg/ml in said medium; and
iii) a microfluidizer capable of maintaining at least 25,000 PSI;
c) using a single pass exposure of said premix to said microfluidizer to create a population of nanoemulsion particles having diameters ranging from between approximately 40-110 nm, wherein said particle diameter remains stable for at least four months.
15. The method of claim 14, further comprising pasteurizing said population of nanoemulsion particles wherein said particle diameters remain stable.
16. The method of claim 14, further comprising freezing said population of nanoemulsion particles wherein said particle diameters remain stable.
17. A method, comprising;
a) providing;
i) a subject refractory to an administered compound at a therapeutically effective amount;
ii) a nanoemulsion comprising a population of particles encapsulating said compound, wherein said particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers;
b) delivering said nanoemulsion to said subjects under conditions such that said compound bioavailability is improved and wherein said compound is therapeutically effective.
18. The method of claim 17, wherein said improved bioavailability comprises pharmacokinetic parameters selected from the group consisting of decreased Tmax, increased Cmax, and increased AUC.
19. The method of claim 17, wherein said delivering comprises a method selected from the group consisting of oral, transdermal, intravenous, intraperitoneal, intramuscular, and subcutaneous.
20. The method of claim 17, wherein said nanoemulsion comprises a plant sterol.
21. The method of claim 17, wherein said nanoemulsion comprises lycopene.
22. A nanoemulsion having bacteria-resistant properties, wherein said nanoemulsion comprises a population of particles encapsulating said compound, wherein said particles having diameters between approximately 10 and approximately 110 nanometers, wherein said nanoemulsion is not contaminated by particles having diameters larger than 110 nanometers.
23. The nanoemulsion of claim 22, wherein said nanoemulsion resists bacterial growth for at least three months.
24. The nanoemulsion of claim 22, wherein said bacterial-resistant properties comprise shear-force induced cell lysis.
25. The nanoemulsion of claim 22, wherein said bacterial-resistant properties comprise an oxidizing environment.
26. The nanoemulsion of claim 22, wherein said nanoemulsion is sterile.
27. A method, comprising:
a) providing;
i) a premix comprising a compound and a liquid dispersion medium; and
ii) a device capable of creating a continuous turbulent flow under high pressure;
b) using said device to create a population of nanoemulsion particles having uniform diameter.
28. The method of claim 27, wherein said dispersion medium is selected from the group consisting of aqueous media and oil-based media.
29. The method of claim 28, wherein said aqueous media is selected from the group consisting of water, saline solution, ringers solution, dextrose, and short chain alcohols.
30. The method of claim 28, wherein said oil-based media is selected from the group consisting of saturated and unsaturated oils from vegetable and marine sources, silicone oils, and mineral oils.
31. The method of claim 27, wherein said compound is selected from the group consisting of a plant sterol, cod liver oil, tocopherol, lecithin, lutein, zeaxanthin, and soy protein.
US11/996,016 2005-07-18 2006-07-11 Compositions and Methods for Making and Using Nanoemulsions Abandoned US20080274195A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/996,016 US20080274195A1 (en) 2005-07-18 2006-07-11 Compositions and Methods for Making and Using Nanoemulsions

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US70022405P 2005-07-18 2005-07-18
PCT/US2006/026918 WO2008010788A2 (en) 2005-07-18 2006-07-11 Compositions and methods for making and using nanoemulsions
US11/996,016 US20080274195A1 (en) 2005-07-18 2006-07-11 Compositions and Methods for Making and Using Nanoemulsions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/026918 A-371-Of-International WO2008010788A2 (en) 2005-07-18 2006-07-11 Compositions and methods for making and using nanoemulsions

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/594,417 Continuation US10016364B2 (en) 2005-07-18 2015-01-12 Compositions and methods for making and using nanoemulsions

Publications (1)

Publication Number Publication Date
US20080274195A1 true US20080274195A1 (en) 2008-11-06

Family

ID=38957224

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/996,016 Abandoned US20080274195A1 (en) 2005-07-18 2006-07-11 Compositions and Methods for Making and Using Nanoemulsions
US14/594,417 Active US10016364B2 (en) 2005-07-18 2015-01-12 Compositions and methods for making and using nanoemulsions

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/594,417 Active US10016364B2 (en) 2005-07-18 2015-01-12 Compositions and methods for making and using nanoemulsions

Country Status (10)

Country Link
US (2) US20080274195A1 (en)
EP (1) EP1919447A4 (en)
JP (2) JP5368793B2 (en)
KR (2) KR20130079645A (en)
CN (2) CN101384247B (en)
AU (1) AU2006346369B2 (en)
BR (1) BRPI0613631A8 (en)
CA (1) CA2618655C (en)
IL (2) IL188666A (en)
WO (1) WO2008010788A2 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080124387A1 (en) * 2006-11-27 2008-05-29 Kapac, Llc Methods and formulations for enhancing the absorption and decreasing the absorption variability of orally administered drugs, vitamins and nutrients
US20090306198A1 (en) * 2005-09-16 2009-12-10 Robert Nicolosi Anti-Oxidant Synergy Formulation Nanoemulsions to Treat Caner
US20100183726A1 (en) * 2006-08-02 2010-07-22 Robert Nicolosi Compositions and methods for treating cancer with dacarbazine nanoemulsions
US20100197785A1 (en) * 2007-07-25 2010-08-05 Epax As Omega-3 fatty acid fortified composition
US20100330135A1 (en) * 2007-07-06 2010-12-30 Fujifilm Corporation Cosmetic composition
US20110206739A1 (en) * 2008-03-28 2011-08-25 University Of Massachusetts Compositions and methods for the preparation of nanoemulsions
US8318181B2 (en) 2005-12-01 2012-11-27 University Of Massachusetts Lowell Botulinum nanoemulsions
US8722131B2 (en) 2010-09-07 2014-05-13 Dsm Nutritional Products Ag Comestible emulsions
US20140271877A1 (en) * 2013-03-15 2014-09-18 Leading Edge Innovations, LLC Substantially surfactant-free, submicron dispersions of hydrophobic agents containing high levels of water miscible solvent
US20150098986A1 (en) * 2013-10-09 2015-04-09 Sky Tree and Camellias Co., Ltd. Method for Producing Elastic Vesicles
US20150342898A1 (en) * 2012-12-21 2015-12-03 Advanced Medical Frontier Co., Ltd. Composition and food or drink
US20160089320A1 (en) * 2014-09-29 2016-03-31 Barrie Tan Non-Synthetic Emulsion-Based Lipid Formulations and Methods of Use
US9486408B2 (en) 2005-12-01 2016-11-08 University Of Massachusetts Lowell Botulinum nanoemulsions
US9486409B2 (en) 2006-12-01 2016-11-08 Anterios, Inc. Peptide nanoparticles and uses therefor
WO2017091462A1 (en) * 2015-11-25 2017-06-01 Pepsico, Inc. Beverage nanoemulstions produced by high shear processing
US20170209372A1 (en) * 2014-08-01 2017-07-27 Hovione International Ltd A Method of Preparing Amorphous Solid Dispersion in Submicron Range by Co-Precipitation
US9724299B2 (en) 2006-12-01 2017-08-08 Anterios, Inc. Amphiphilic entity nanoparticles
US10016451B2 (en) 2007-05-31 2018-07-10 Anterios, Inc. Nucleic acid nanoparticles and uses therefor
US10016364B2 (en) 2005-07-18 2018-07-10 University Of Massachusetts Lowell Compositions and methods for making and using nanoemulsions
US10206417B1 (en) * 2018-06-07 2019-02-19 King Saud University Guava seed (Psidium guajava) nanoparticles as antibacterial agent
US20190110986A1 (en) * 2015-10-08 2019-04-18 Nof Corporation O/w type emulsion
US10531674B2 (en) 2013-03-15 2020-01-14 Leading Edge Innovations, LLC Compositions having an oil-in-water dispersion of submicron particles to enhance foods and beverages
US10596117B1 (en) * 2014-12-31 2020-03-24 Eric Morrison Lipoleosomes as carriers for aromatic amide anesthetic compounds
CN112042928A (en) * 2020-08-31 2020-12-08 华南理工大学 Method for synergistically and efficiently preparing protein-based nano emulsion by using polyhydroxy alcohol as molecular chaperone and prepared protein-based nano emulsion
US20220054414A1 (en) * 2020-08-19 2022-02-24 Readymix Foods Corp. Nanoemulsion Compositions Comprising Saponins for Increasing Bioavailability
US11311496B2 (en) 2016-11-21 2022-04-26 Eirion Therapeutics, Inc. Transdermal delivery of large agents
US11465107B2 (en) * 2019-01-31 2022-10-11 Hong Ngoc Thi Dang Process for producing a nano omega-3 microemulsion system
US11622947B2 (en) 2019-05-31 2023-04-11 American River Nutrition, Llc Compositions comprising quillaja extract and methods of preparations and use thereof

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9000040B2 (en) 2004-09-28 2015-04-07 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
EP1811933B1 (en) 2004-09-28 2016-03-23 Atrium Medical Corporation Barrier layer
US9012506B2 (en) 2004-09-28 2015-04-21 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US20060088596A1 (en) 2004-09-28 2006-04-27 Atrium Medical Corporation Solubilizing a drug for use in a coating
US9801982B2 (en) 2004-09-28 2017-10-31 Atrium Medical Corporation Implantable barrier device
US9427423B2 (en) 2009-03-10 2016-08-30 Atrium Medical Corporation Fatty-acid based particles
US9278161B2 (en) 2005-09-28 2016-03-08 Atrium Medical Corporation Tissue-separating fatty acid adhesion barrier
WO2008057328A2 (en) 2006-11-06 2008-05-15 Atrium Medical Corporation Tissue separating device with reinforced support for anchoring mechanisms
US9492596B2 (en) 2006-11-06 2016-11-15 Atrium Medical Corporation Barrier layer with underlying medical device and one or more reinforcing support structures
US8357639B2 (en) 2007-07-03 2013-01-22 Baker Hughes Incorporated Nanoemulsions
US8282977B2 (en) 2008-03-20 2012-10-09 Virun, Inc. Compositions containing non-polar compounds
US20110038910A1 (en) 2009-08-11 2011-02-17 Atrium Medical Corporation Anti-infective antimicrobial-containing biomaterials
US20110045050A1 (en) * 2009-08-24 2011-02-24 Atrium Medical Corporation Nanoemulsion formulations for direct delivery
ITBS20100017A1 (en) * 2010-02-03 2011-08-04 Francesco Palladini PROCEDURE FOR THE PRODUCTION OF A FOOD AND SUPPLEMENTARY SUPPLEMENTER SO OBTAINED
EP2593141B1 (en) 2010-07-16 2018-07-04 Atrium Medical Corporation Composition and methods for altering the rate of hydrolysis of cured oil-based materials
EP2667945A1 (en) 2011-01-24 2013-12-04 Anterios, Inc. Oil compositions
BR112013018918A2 (en) 2011-01-24 2016-10-04 Anterios Inc nanoparticle compositions
CN106924216A (en) 2011-01-24 2017-07-07 安特里奥公司 Nanoparticulate compositions, its preparation and their purposes
US9867880B2 (en) 2012-06-13 2018-01-16 Atrium Medical Corporation Cured oil-hydrogel biomaterial compositions for controlled drug delivery
WO2015057751A1 (en) * 2013-10-14 2015-04-23 Nanosphere Health Sciences, Llc Nanoparticle compositions and methods as carriers of nutraceutical factors across cell membranes and biological barriers
JP2017506671A (en) * 2014-02-14 2017-03-09 ホアン,ジンジュン Composition of nanoemulsion delivery system
WO2016100228A2 (en) 2014-12-15 2016-06-23 Nanosphere Health Sciences, Llc Methods of treating inflammatory disorders and global inflammation with compositions comprising phospholipid nanoparticle encapsulations of nsaids
WO2016144376A1 (en) 2015-03-10 2016-09-15 Nanosphere Health Sciences, Llc Lipid nanoparticle compositions and methods as carriers of cannabinoids in standardized precision-metered dosage forms
CN106551902B (en) * 2015-09-17 2020-07-03 阿赖耶识(上海)生物技术有限公司 High-stability non-vesicular nanoparticles and application thereof in treating microbial infection
GB201618350D0 (en) * 2016-10-31 2016-12-14 Ip Science Ltd Methods and compositions comprising carotenoids
CN108576778A (en) * 2018-02-06 2018-09-28 华南农业大学 A kind of food grade excipients nanoemulsions and the preparation method and application thereof
SG11202106144VA (en) 2018-12-11 2021-07-29 Disruption Labs Inc Compositions for the delivery of therapeutic agents and methods of use and making thereof
CN111296730A (en) * 2020-03-06 2020-06-19 广州市食品工业研究所有限公司 Lutein nanoemulsion with good stability and preparation method thereof
CN111544387B (en) * 2020-04-18 2021-11-02 内蒙古医科大学 Compound quercetin antibacterial nanoemulsion and preparation method thereof
WO2024006539A1 (en) * 2022-07-01 2024-01-04 Vitakey Inc. Nutraceutical particles

Citations (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4533254A (en) * 1981-04-17 1985-08-06 Biotechnology Development Corporation Apparatus for forming emulsions
US4908154A (en) * 1981-04-17 1990-03-13 Biotechnology Development Corporation Method of forming a microemulsion
US5152923A (en) * 1989-06-26 1992-10-06 Hans Georg Weder Process for the production of a nanoemulsion of oil particles in an aqueous phase
US5401243A (en) * 1990-08-21 1995-03-28 Associated Synapse Biologics Controlled administration of chemodenervating pharmaceuticals
US5502045A (en) * 1991-05-03 1996-03-26 Raision Tehtaat Oy Ab Use of a stanol fatty acid ester for reducing serum cholesterol level
US5510118A (en) * 1995-02-14 1996-04-23 Nanosystems Llc Process for preparing therapeutic compositions containing nanoparticles
US5576016A (en) * 1993-05-18 1996-11-19 Pharmos Corporation Solid fat nanoemulsions as drug delivery vehicles
US5629021A (en) * 1995-01-31 1997-05-13 Novavax, Inc. Micellar nanoparticles
US5651991A (en) * 1987-10-28 1997-07-29 Nippon Shinyaku Co. Ltd. Drug carriers
US5670484A (en) * 1994-05-09 1997-09-23 Binder; William J. Method for treatment of skin lesions associated with cutaneous cell-proliferative disorders
US5672358A (en) * 1994-06-21 1997-09-30 Ascent Pharmaceuticals, Inc. Controlled release aqueous emulsion
US5753241A (en) * 1995-02-27 1998-05-19 L'oreal Transparent nanoemulsion less than 100 NM based on fluid non-ionic amphiphilic lipids and use in cosmetic or in dermopharmaceuticals
US5766605A (en) * 1994-04-15 1998-06-16 Mount Sinai School Of Medicine Of The City University Of New York Treatment of autonomic nerve dysfunction with botulinum toxin
US5858410A (en) * 1994-11-11 1999-01-12 Medac Gesellschaft Fur Klinische Spezialpraparate Pharmaceutical nanosuspensions for medicament administration as systems with increased saturation solubility and rate of solution
US5925341A (en) * 1997-03-18 1999-07-20 L'oreal Nanoemulsion based on nonionic amphiphilic lipids and aminated silicones and uses
US5932562A (en) * 1998-05-26 1999-08-03 Washington University Sitostanol formulation to reduce cholesterol absorption and method for preparing and use of same
US5994414A (en) * 1997-04-28 1999-11-30 Avon Products, Inc. Water-thin emulsion formed by high pressure homogenization process
US6007856A (en) * 1997-08-08 1999-12-28 The Procter & Gamble Company Oil-in-water dispersions of β-carotene and other carotenoids stable against oxidation prepared from water-dispersible beadlets having high concentrations of carotenoid
US6039936A (en) * 1996-11-15 2000-03-21 L'oreal Nanoemulsion based on non-ionic and cationic amphiphilic lipids and uses thereof
US6224853B1 (en) * 1997-04-22 2001-05-01 Woolcombers Group Plc Aqueous compositions comprising a lipid and a lanolin-derived surfactant, and their use
US6265180B1 (en) * 1998-03-30 2001-07-24 Mibelle Ag Cosmetics Nanoemulsions for delivering lipophilic substances into cells
US6274150B1 (en) * 1998-12-23 2001-08-14 L'oreal Nanoemulsion based on phosphoric acid fatty acid esters and its uses in the cosmetics, dermatological, pharmaceutical, and/or ophthalmological fields
US6312708B1 (en) * 2000-06-02 2001-11-06 Allergan Sales, Inc. Botulinum toxin implant
US6358917B1 (en) * 1999-08-24 2002-03-19 Jean D. A. Carruthers Cosmetic use of botulinum toxin for treatment of downturned mouth
US20020048596A1 (en) * 1994-12-30 2002-04-25 Gregor Cevc Preparation for the transport of an active substance across barriers
US6387411B2 (en) * 1998-11-30 2002-05-14 Mcneil-Ppc, Inc. Method for producing dispersible sterol and stanol compounds
US20020098215A1 (en) * 2000-01-21 2002-07-25 Veronique Douin Nanoemulsions comprising at least one amphiphilic lipid, at least one oil, and at least one nonionic polymer, and uses thereof
US6429189B1 (en) * 1999-12-10 2002-08-06 Botulinum Toxin Research Associates, Inc. Cytotoxin (non-neurotoxin) for the treatment of human headache disorders and inflammatory diseases
US20020107199A1 (en) * 2000-12-05 2002-08-08 Allergan Sales, Inc. Methods of administering botulinum toxin
US20020155084A1 (en) * 2000-06-02 2002-10-24 The Regents Of The University Of The Michigan Nanoemulsion formulations
US20030072801A1 (en) * 2001-06-22 2003-04-17 Pfizer Inc. Pharmaceutical compositions comprising drug and concentration-enhancing polymers
US6573241B1 (en) * 1998-12-10 2003-06-03 BioteCon Gesellschaft für bio-technologische Entwicklung und Consulting GmbH Therapeutic agent for the suppression of snoring noises
US20030108597A1 (en) * 2001-08-13 2003-06-12 Chancellor Michael B. Application of lipid vehicles and use for drug delivery
US6589588B1 (en) * 1998-05-06 2003-07-08 Raisio Benecol Oy Phytosterol compositions
US6623780B1 (en) * 2002-03-26 2003-09-23 Cargill, Inc. Aqueous dispersible sterol product
US6632440B1 (en) * 1998-08-25 2003-10-14 Health Protection Agency Methods and compounds for the treatment of mucus hypersecretion
US20030194412A1 (en) * 2001-06-05 2003-10-16 The Regents Of The University Of Michigan Nanoemulsion vaccines
US20030206955A1 (en) * 2000-05-22 2003-11-06 L'oreal Nanoemulsions, compositions comprising such nanoemulsions, and methods of using such nanoemulsions
US20040003324A1 (en) * 2002-06-29 2004-01-01 Richard Uhlig Handling faults associated with operation of guest software in the virtual-machine architecture
US20040005370A1 (en) * 2000-07-13 2004-01-08 Lionel Breton Composition, in particular cosmetic, containing dhea and/or a chemical or biological precursor or derivative thereof , and a metalloproteinase inhibitors
US20040009936A1 (en) * 1999-05-03 2004-01-15 Tang De-Chu C. Vaccine and drug delivery by topical application of vectors and vector extracts
US20040009180A1 (en) * 2002-07-11 2004-01-15 Allergan, Inc. Transdermal botulinum toxin compositions
US6688311B2 (en) * 2002-03-14 2004-02-10 Allergan, Inc. Method for determining effect of a clostridial toxin upon a muscle
US20040033202A1 (en) * 2002-06-10 2004-02-19 Elan Pharma International, Ltd. Nanoparticulate sterol formulations and novel sterol combinations
US20040037853A1 (en) * 2002-05-28 2004-02-26 Gary Borodic Composition for therapeutic and cosmetic botulinum toxin
US20040048836A1 (en) * 2001-03-23 2004-03-11 Wilmott James M Method for preparing high pressure/ high shear dispersions containing physiologically active ingredients
US6720001B2 (en) * 1999-10-18 2004-04-13 Lipocine, Inc. Emulsion compositions for polyfunctional active ingredients
US20040081688A1 (en) * 2000-12-27 2004-04-29 Del Curto Maria Dorly Amphiphilic lipid nanoparticles for peptide and/or protein incorporation
US20040115727A1 (en) * 2002-12-11 2004-06-17 Allergan, Inc., A Corporation Evolved clostridial toxins with altered protease specificity
US20040115159A1 (en) * 2002-03-29 2004-06-17 Tadlock Charles C Novel nanoemulsions
US20040126397A1 (en) * 1993-12-28 2004-07-01 Allergan, Inc. Use of the neurotoxic component of a botulinum toxin for treating various disorders and conditions and associated pain
US20040127661A1 (en) * 2002-09-12 2004-07-01 Harald Kaspar Fluoroelastomers having low temperature characteristics and solvent resistance
US20040151741A1 (en) * 2002-12-20 2004-08-05 Gary Borodic Pharmaceutical botulinum toxin compositions
US20040229038A1 (en) * 2003-03-03 2004-11-18 Elan Pharma International Ltd. Nanoparticulate meloxicam formulations
US6861066B2 (en) * 2002-03-11 2005-03-01 Health Plus International Inc. Method for the delivery of a biologically active agent
US20050048088A1 (en) * 2003-08-28 2005-03-03 Fred Zulli Compositions comprising at least two nanoemulsions
US6869610B2 (en) * 2000-04-14 2005-03-22 Allergan Sales, Inc. Pain treatment by peripheral administration of a neurotoxin
US20050074466A1 (en) * 2001-07-27 2005-04-07 Suskind Dana L. Botulinum toxin in the treatment or prevention of acne
US20050079131A1 (en) * 2003-08-08 2005-04-14 Lanza Gregory M. Emulsion particles for imaging and therapy and methods of use thereof
US20050096340A1 (en) * 2003-10-29 2005-05-05 Yuehua Zhang Tocopherol-modified therapeutic drug compounds
US6902737B2 (en) * 2001-01-18 2005-06-07 L'oreal Translucent nanoemulsion, production method, and uses thereof in the cosmetic, dermatological and/or ophthalmological fields
US20050123897A1 (en) * 1999-07-05 2005-06-09 Idea Ag Method for the improvement of transport across adaptable semi-permeable barriers
US20050142150A1 (en) * 1991-09-24 2005-06-30 Allergan, Inc. Botulinum toxin formulations
US20050184275A1 (en) * 2004-02-23 2005-08-25 The Texas A&M University System Antioxidant compositions and methods of use thereof
US6939852B2 (en) * 1991-09-24 2005-09-06 Allergan, Inc. Methods and compositions for the treatment of cerebral palsy
US20050196414A1 (en) * 2004-03-03 2005-09-08 Essentia Biosystems, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US20050208083A1 (en) * 2003-06-04 2005-09-22 Nanobio Corporation Compositions for inactivating pathogenic microorganisms, methods of making the compositons, and methods of use thereof
US20050214325A1 (en) * 2004-03-26 2005-09-29 Vvii Newco 2003, Inc. Compositions and methods to increase the effect of a neurotoxin treatment
US20050249686A1 (en) * 2004-04-27 2005-11-10 Francoise Pataut Cosmetic composition free of water comprising fatty acid esters, petrolatum oil and non-ionic polymers
US20060018931A1 (en) * 2004-07-26 2006-01-26 Taylor Harold V Therapeutic composition with a botulinum neurotoxin
US7001602B2 (en) * 1997-07-15 2006-02-21 The Regents Of The University Of Colorado Use of botulinum toxin therapy for urinary incontinence and related disorders
US20060057165A1 (en) * 2004-09-10 2006-03-16 Dimitrios Dimitrakoudis Clostridium botulinum toxin formulation and method for reducing weight
US20060073208A1 (en) * 2004-10-01 2006-04-06 Allergan, Inc. Cosmetic neurotoxin compositions and methods
US20060093624A1 (en) * 1991-09-24 2006-05-04 Allergan, Inc. Botulinum toxin for treating muscle contracture
US20060153877A1 (en) * 2003-06-20 2006-07-13 Shunji Kozaki Remedies for dissease with hypermyotonia
US20060153876A1 (en) * 2003-02-24 2006-07-13 Ira Sanders Cell membrane translocation of regulated snare inhibitors, compositions therefor, and methods for treatment of disease
US20060165657A1 (en) * 2003-02-05 2006-07-27 Paul Bernasconi Method for delivery of cosmetic by topical application
US20060182767A1 (en) * 2002-05-28 2006-08-17 Borodic Gary E High-potency botulinum toxin formulations
US20070036831A1 (en) * 2005-08-09 2007-02-15 Nanobio Corporation Nanoemulsion compositions having anti-inflammatory activity
US20070116723A1 (en) * 2001-12-18 2007-05-24 Coleman William P Iii Topically applied clostridium botulinum toxin compositions and treatment methods
US7228259B2 (en) * 2004-06-30 2007-06-05 Lucent Technologies Inc. Method and apparatus for structure-preserving reduced-order modeling
US20070148194A1 (en) * 2005-11-29 2007-06-28 Amiji Mansoor M Novel nanoemulsion formulations
US20070178121A1 (en) * 2006-01-27 2007-08-02 Allergan, Inc. Methods for enhancing skin treatments
US20100040883A1 (en) * 2001-12-19 2010-02-18 Mccarthy Stephen P Polysaccharide-containing block copolymer particles and uses thereof
US20100183726A1 (en) * 2006-08-02 2010-07-22 Robert Nicolosi Compositions and methods for treating cancer with dacarbazine nanoemulsions
US20110206736A1 (en) * 2008-09-23 2011-08-25 Thomas Jefferson University Cancer Vaccines Against Mucosal Antigens and Methods of Making and Using the Same

Family Cites Families (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH587021A5 (en) * 1974-06-07 1977-04-29 Nestle Sa
US5554372A (en) * 1986-09-22 1996-09-10 Emory University Methods and vaccines comprising surface-active copolymers
JPH0798740B2 (en) * 1987-10-28 1995-10-25 日本新薬株式会社 Drug carrier
IL88076A (en) 1987-10-28 1993-01-14 Nippon Shinyaku Co Ltd Fat emulsions as drug carriers
US6165500A (en) 1990-08-24 2000-12-26 Idea Ag Preparation for the application of agents in mini-droplets
US5652274A (en) 1991-03-01 1997-07-29 Martin; Alain Therapeutic-wound healing compositions and methods for preparing and using same
EP0572080B1 (en) 1992-05-29 1995-11-15 Quest International B.V. Aqueous perfume oil microemulsions
EP0571677A1 (en) 1992-05-29 1993-12-01 Unilever Plc Aqueous parfume oil microemulsions
US5302401A (en) * 1992-12-09 1994-04-12 Sterling Winthrop Inc. Method to reduce particle size growth during lyophilization
CA2091152C (en) * 1993-03-05 2005-05-03 Kirsten Westesen Solid lipid particles, particles of bioactive agents and methods for the manfuacture and use thereof
JP2867208B2 (en) * 1993-07-27 1999-03-08 光洋精工株式会社 Clutch release bearing
US6974578B1 (en) 1993-12-28 2005-12-13 Allergan, Inc. Method for treating secretions and glands using botulinum toxin
EP0746318A1 (en) 1994-02-25 1996-12-11 Takeda Chemical Industries, Ltd. Injectable emulsions containing antifungal triazole derivatives
JPH07285863A (en) * 1994-02-25 1995-10-31 Takeda Chem Ind Ltd Composition for injection and its production
ES2078190B1 (en) 1994-05-20 1996-08-01 Cusi Lab PROCEDURE FOR THE COATING OF GOTICLES OR PARTICLES OF NANOMETRIC SIZE.
US5843334A (en) 1994-06-20 1998-12-01 Nippon Shinyaku Co., Ltd. Method of producing emulsions and an emulsification apparatus
US6835395B1 (en) 1997-05-14 2004-12-28 The University Of British Columbia Composition containing small multilamellar oligodeoxynucleotide-containing lipid vesicles
US6287591B1 (en) 1997-05-14 2001-09-11 Inex Pharmaceuticals Corp. Charged therapeutic agents encapsulated in lipid particles containing four lipid components
JPH1144594A (en) * 1997-07-25 1999-02-16 Yamatake Honeywell Co Ltd Pressure detector
US5993852A (en) * 1997-08-29 1999-11-30 Pharmaderm Laboratories Ltd. Biphasic lipid vesicle composition for transdermal administration of an immunogen
CA2322805C (en) 1998-03-05 2005-09-13 Nippon Shinyaku Co., Ltd. Fat emulsions for inhalational administration
TR200101790T2 (en) 1998-12-23 2001-10-22 Idea Ag. Formula developed for uncommon in vivo topical applications
FR2788007B1 (en) 1999-01-05 2001-02-09 Oreal NANOEMULSION BASED ON BLOCK COPOLYMERS OF ETHYLENE OXIDE AND PROPYLENE OXIDE, AND ITS USES IN THE COSMETIC, DERMATOLOGICAL AND / OR OPHTHALMOLOGICAL FIELDS
JP2003506398A (en) 1999-08-04 2003-02-18 イデア アクチェンゲゼルシャフト Periodic structures containing lipids, polyelectrolytes, and structure-inducing soluble low-valent linkers, and methods for their biological use
AU2001245947A1 (en) 2000-03-23 2001-10-03 Collaborative Technologies, Inc Method for preparing high pressure/high shear dispersions containing physiologically active ingredients
US6471968B1 (en) 2000-05-12 2002-10-29 Regents Of The University Of Michigan Multifunctional nanodevice platform
US6670322B2 (en) 2000-06-01 2003-12-30 Wisconsin Alumni Research Foundation Method of targeting pharmaceuticals to motor neurons
US20040033241A1 (en) 2000-06-02 2004-02-19 Allergan, Inc. Controlled release botulinum toxin system
US20040220100A1 (en) 2000-07-21 2004-11-04 Essentia Biosystems, Inc. Multi-component biological transport systems
FI20010115A0 (en) 2001-01-18 2001-01-18 Orion Corp A process for preparing nanoparticles
AU2002258644B2 (en) * 2001-03-30 2006-12-14 Color Access, Inc. Novel nanoemulsions
KR100463167B1 (en) * 2001-04-13 2004-12-23 주식회사 태평양 Percutaneous Controlled Releasing Material Using Nano-sized Polymeric Particles and External Application Agent Containing the Same
US20030224020A1 (en) 2002-05-31 2003-12-04 Zabudkin Alexander F. Pharmaceutical preparation of botulinum neurotoxin, methods of synthesis and methods of clinical use
US7329688B2 (en) * 2002-12-17 2008-02-12 Soft Gel Technologies, Inc. Natural vitamin E compositions with superior antioxidant potency
CA2513064C (en) 2003-01-31 2009-11-10 Elan Pharma International, Ltd. Nanoparticulate topiramate formulations
US20040247623A1 (en) 2003-03-24 2004-12-09 Roger Cady Method and article for treatment of sensory neuron related disorders through transdermal application of botulinum toxin
JP2006522090A (en) * 2003-04-04 2006-09-28 ファイザー・プロダクツ・インク Microfluidized oil-in-water emulsion and vaccine composition
WO2005027872A2 (en) * 2003-06-04 2005-03-31 Nanobio Corporation Compositions for inactivating pathogenic microorganisms, methods of making the compositions, and methods of use thereof
US20060151899A1 (en) 2003-08-06 2006-07-13 Akira Kato Process for producing drug ultramicroparticle and apparatus therefor
EP1510206A1 (en) * 2003-08-29 2005-03-02 Novagali Pharma SA Self-nanoemulsifying oily formulation for the administration of poorly water-soluble drugs
EP1547674A1 (en) 2003-12-23 2005-06-29 MediGene Oncology GmbH Method for producing colloidal nanoparticles
US6974579B2 (en) 2004-01-08 2005-12-13 Allergan, Inc. Methods for treating vascular disorders
EP1713446B1 (en) 2004-01-23 2010-12-01 Camurus Ab Ternary non-lamellar lipid compositions
TW200603843A (en) 2004-04-20 2006-02-01 Technology Dev Company Ltd Tissue enhancement implant and method
US7591806B2 (en) 2004-05-18 2009-09-22 Bai Xu High-aspect-ratio microdevices and methods for transdermal delivery and sampling of active substances
GB2416122A (en) 2004-07-12 2006-01-18 Ipsen Ltd Botulinum neurotoxin composition
CA2584475A1 (en) 2004-11-12 2006-05-18 Idea Ag Extended surface aggregates in the treatment of skin conditions
PT1853303E (en) 2005-02-14 2016-03-23 Dpm Therapeutics Corp Stabilized compositions for topical administration and methods of making same
CA2494473C (en) 2005-02-14 2007-06-26 Dpm Therapeutics Corp. Stabilized protein compositions for topical administration and methods of making same
EP1861112A4 (en) 2005-03-03 2009-07-22 Revance Therapeutics Inc Compositions and methods for topical application and transdermal delivery of botulinum toxins
EP2248534A1 (en) 2005-06-14 2010-11-10 Botulinum Toxin Research Associates, Inc. Botulinum toxin and the treatment of primary disorders of mood and affect
JP5368793B2 (en) 2005-07-18 2013-12-18 ユニバーシティ オブ マサチューセッツ ロウエル Compositions and methods for making and using nanoemulsions
US20090306198A1 (en) 2005-09-16 2009-12-10 Robert Nicolosi Anti-Oxidant Synergy Formulation Nanoemulsions to Treat Caner
US8137677B2 (en) 2005-10-06 2012-03-20 Allergan, Inc. Non-protein stabilized clostridial toxin pharmaceutical compositions
US9486408B2 (en) 2005-12-01 2016-11-08 University Of Massachusetts Lowell Botulinum nanoemulsions
DK3295955T3 (en) 2005-12-01 2021-07-05 Univ Massachusetts Lowell BOTULINUM NANOEMULSIONS
WO2007103555A2 (en) 2006-03-08 2007-09-13 Nuviance, Inc. Transdermal drug delivery compositions and topical compositions for application on the skin
US20110206739A1 (en) 2008-03-28 2011-08-25 University Of Massachusetts Compositions and methods for the preparation of nanoemulsions

Patent Citations (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4533254A (en) * 1981-04-17 1985-08-06 Biotechnology Development Corporation Apparatus for forming emulsions
US4908154A (en) * 1981-04-17 1990-03-13 Biotechnology Development Corporation Method of forming a microemulsion
US5651991A (en) * 1987-10-28 1997-07-29 Nippon Shinyaku Co. Ltd. Drug carriers
US5152923A (en) * 1989-06-26 1992-10-06 Hans Georg Weder Process for the production of a nanoemulsion of oil particles in an aqueous phase
US5401243A (en) * 1990-08-21 1995-03-28 Associated Synapse Biologics Controlled administration of chemodenervating pharmaceuticals
US5502045A (en) * 1991-05-03 1996-03-26 Raision Tehtaat Oy Ab Use of a stanol fatty acid ester for reducing serum cholesterol level
US7384918B2 (en) * 1991-09-24 2008-06-10 Allergan, Inc. Botulinum toxin for treating muscle contracture
US20050142150A1 (en) * 1991-09-24 2005-06-30 Allergan, Inc. Botulinum toxin formulations
US6939852B2 (en) * 1991-09-24 2005-09-06 Allergan, Inc. Methods and compositions for the treatment of cerebral palsy
US20060093624A1 (en) * 1991-09-24 2006-05-04 Allergan, Inc. Botulinum toxin for treating muscle contracture
US5576016A (en) * 1993-05-18 1996-11-19 Pharmos Corporation Solid fat nanoemulsions as drug delivery vehicles
US20040126397A1 (en) * 1993-12-28 2004-07-01 Allergan, Inc. Use of the neurotoxic component of a botulinum toxin for treating various disorders and conditions and associated pain
US5766605A (en) * 1994-04-15 1998-06-16 Mount Sinai School Of Medicine Of The City University Of New York Treatment of autonomic nerve dysfunction with botulinum toxin
US5670484A (en) * 1994-05-09 1997-09-23 Binder; William J. Method for treatment of skin lesions associated with cutaneous cell-proliferative disorders
US5672358A (en) * 1994-06-21 1997-09-30 Ascent Pharmaceuticals, Inc. Controlled release aqueous emulsion
US5858410A (en) * 1994-11-11 1999-01-12 Medac Gesellschaft Fur Klinische Spezialpraparate Pharmaceutical nanosuspensions for medicament administration as systems with increased saturation solubility and rate of solution
US20020048596A1 (en) * 1994-12-30 2002-04-25 Gregor Cevc Preparation for the transport of an active substance across barriers
US5629021A (en) * 1995-01-31 1997-05-13 Novavax, Inc. Micellar nanoparticles
US5510118A (en) * 1995-02-14 1996-04-23 Nanosystems Llc Process for preparing therapeutic compositions containing nanoparticles
US5753241A (en) * 1995-02-27 1998-05-19 L'oreal Transparent nanoemulsion less than 100 NM based on fluid non-ionic amphiphilic lipids and use in cosmetic or in dermopharmaceuticals
US6039936A (en) * 1996-11-15 2000-03-21 L'oreal Nanoemulsion based on non-ionic and cationic amphiphilic lipids and uses thereof
US5925341A (en) * 1997-03-18 1999-07-20 L'oreal Nanoemulsion based on nonionic amphiphilic lipids and aminated silicones and uses
US6224853B1 (en) * 1997-04-22 2001-05-01 Woolcombers Group Plc Aqueous compositions comprising a lipid and a lanolin-derived surfactant, and their use
US5994414A (en) * 1997-04-28 1999-11-30 Avon Products, Inc. Water-thin emulsion formed by high pressure homogenization process
US7001602B2 (en) * 1997-07-15 2006-02-21 The Regents Of The University Of Colorado Use of botulinum toxin therapy for urinary incontinence and related disorders
US6007856A (en) * 1997-08-08 1999-12-28 The Procter & Gamble Company Oil-in-water dispersions of β-carotene and other carotenoids stable against oxidation prepared from water-dispersible beadlets having high concentrations of carotenoid
US6265180B1 (en) * 1998-03-30 2001-07-24 Mibelle Ag Cosmetics Nanoemulsions for delivering lipophilic substances into cells
US6558941B2 (en) * 1998-03-30 2003-05-06 Mibelle Ag Cosmetics Delivering lipophilic substances into cells using nanoemulsions
US6589588B1 (en) * 1998-05-06 2003-07-08 Raisio Benecol Oy Phytosterol compositions
US5932562A (en) * 1998-05-26 1999-08-03 Washington University Sitostanol formulation to reduce cholesterol absorption and method for preparing and use of same
US6632440B1 (en) * 1998-08-25 2003-10-14 Health Protection Agency Methods and compounds for the treatment of mucus hypersecretion
US6387411B2 (en) * 1998-11-30 2002-05-14 Mcneil-Ppc, Inc. Method for producing dispersible sterol and stanol compounds
US6573241B1 (en) * 1998-12-10 2003-06-03 BioteCon Gesellschaft für bio-technologische Entwicklung und Consulting GmbH Therapeutic agent for the suppression of snoring noises
US6274150B1 (en) * 1998-12-23 2001-08-14 L'oreal Nanoemulsion based on phosphoric acid fatty acid esters and its uses in the cosmetics, dermatological, pharmaceutical, and/or ophthalmological fields
US20040009936A1 (en) * 1999-05-03 2004-01-15 Tang De-Chu C. Vaccine and drug delivery by topical application of vectors and vector extracts
US20050123897A1 (en) * 1999-07-05 2005-06-09 Idea Ag Method for the improvement of transport across adaptable semi-permeable barriers
USRE39086E1 (en) * 1999-08-24 2006-05-02 Allergan, Inc. Cosmetic use of botulinum toxin for treatment of downturned mouth
US6358917B1 (en) * 1999-08-24 2002-03-19 Jean D. A. Carruthers Cosmetic use of botulinum toxin for treatment of downturned mouth
US6720001B2 (en) * 1999-10-18 2004-04-13 Lipocine, Inc. Emulsion compositions for polyfunctional active ingredients
US6429189B1 (en) * 1999-12-10 2002-08-06 Botulinum Toxin Research Associates, Inc. Cytotoxin (non-neurotoxin) for the treatment of human headache disorders and inflammatory diseases
US20050226842A1 (en) * 2000-01-21 2005-10-13 L'oreal S.A. Nanoemulsions comprising at least one amphiphilic lipid, at least one oil, and at least one nonionic polymer, and uses thereof
US20030138465A9 (en) * 2000-01-21 2003-07-24 Veronique Douin Nanoemulsions comprising at least one amphiphilic lipid, at least one oil, and at least one nonionic polymer, and uses thereof
US20020098215A1 (en) * 2000-01-21 2002-07-25 Veronique Douin Nanoemulsions comprising at least one amphiphilic lipid, at least one oil, and at least one nonionic polymer, and uses thereof
US6869610B2 (en) * 2000-04-14 2005-03-22 Allergan Sales, Inc. Pain treatment by peripheral administration of a neurotoxin
US20030206955A1 (en) * 2000-05-22 2003-11-06 L'oreal Nanoemulsions, compositions comprising such nanoemulsions, and methods of using such nanoemulsions
US20020155084A1 (en) * 2000-06-02 2002-10-24 The Regents Of The University Of The Michigan Nanoemulsion formulations
US6312708B1 (en) * 2000-06-02 2001-11-06 Allergan Sales, Inc. Botulinum toxin implant
US20040005370A1 (en) * 2000-07-13 2004-01-08 Lionel Breton Composition, in particular cosmetic, containing dhea and/or a chemical or biological precursor or derivative thereof , and a metalloproteinase inhibitors
US20020107199A1 (en) * 2000-12-05 2002-08-08 Allergan Sales, Inc. Methods of administering botulinum toxin
US7255865B2 (en) * 2000-12-05 2007-08-14 Allergan, Inc. Methods of administering botulinum toxin
US20040081688A1 (en) * 2000-12-27 2004-04-29 Del Curto Maria Dorly Amphiphilic lipid nanoparticles for peptide and/or protein incorporation
US6902737B2 (en) * 2001-01-18 2005-06-07 L'oreal Translucent nanoemulsion, production method, and uses thereof in the cosmetic, dermatological and/or ophthalmological fields
US20040048836A1 (en) * 2001-03-23 2004-03-11 Wilmott James M Method for preparing high pressure/ high shear dispersions containing physiologically active ingredients
US20030194412A1 (en) * 2001-06-05 2003-10-16 The Regents Of The University Of Michigan Nanoemulsion vaccines
US20030072801A1 (en) * 2001-06-22 2003-04-17 Pfizer Inc. Pharmaceutical compositions comprising drug and concentration-enhancing polymers
US7226605B2 (en) * 2001-07-27 2007-06-05 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Botulinum toxin in the treatment or prevention of acne
US20050074466A1 (en) * 2001-07-27 2005-04-07 Suskind Dana L. Botulinum toxin in the treatment or prevention of acne
US20030108597A1 (en) * 2001-08-13 2003-06-12 Chancellor Michael B. Application of lipid vehicles and use for drug delivery
US7507419B2 (en) * 2001-12-18 2009-03-24 Coleman Iii William P Topically applied Clostridium botulinum toxin compositions and treatment methods
US20070116723A1 (en) * 2001-12-18 2007-05-24 Coleman William P Iii Topically applied clostridium botulinum toxin compositions and treatment methods
US20100040883A1 (en) * 2001-12-19 2010-02-18 Mccarthy Stephen P Polysaccharide-containing block copolymer particles and uses thereof
US20110020227A1 (en) * 2001-12-19 2011-01-27 University Of Massachusetts Polysaccharide-containing block copolmer particles and uses thereof
US7763663B2 (en) * 2001-12-19 2010-07-27 University Of Massachusetts Polysaccharide-containing block copolymer particles and uses thereof
US6861066B2 (en) * 2002-03-11 2005-03-01 Health Plus International Inc. Method for the delivery of a biologically active agent
US20050147688A1 (en) * 2002-03-11 2005-07-07 Russell Van De Casteele Method for the delivery of a biologically active agent
US6688311B2 (en) * 2002-03-14 2004-02-10 Allergan, Inc. Method for determining effect of a clostridial toxin upon a muscle
US6623780B1 (en) * 2002-03-26 2003-09-23 Cargill, Inc. Aqueous dispersible sterol product
US20040115159A1 (en) * 2002-03-29 2004-06-17 Tadlock Charles C Novel nanoemulsions
US20040037853A1 (en) * 2002-05-28 2004-02-26 Gary Borodic Composition for therapeutic and cosmetic botulinum toxin
US20060182767A1 (en) * 2002-05-28 2006-08-17 Borodic Gary E High-potency botulinum toxin formulations
US20040033202A1 (en) * 2002-06-10 2004-02-19 Elan Pharma International, Ltd. Nanoparticulate sterol formulations and novel sterol combinations
US20040003324A1 (en) * 2002-06-29 2004-01-01 Richard Uhlig Handling faults associated with operation of guest software in the virtual-machine architecture
US20050074461A1 (en) * 2002-07-11 2005-04-07 Stephen Donovan Transdermal botulinum toxin compositions
US20050175636A1 (en) * 2002-07-11 2005-08-11 Stephen Donovan Transdermal patch for botulinum toxin administration
US20040009180A1 (en) * 2002-07-11 2004-01-15 Allergan, Inc. Transdermal botulinum toxin compositions
US20060188525A1 (en) * 2002-07-11 2006-08-24 Allergan, Inc. Transdermal botulinum toxin administration for the treatment of spastic muscle pain
US20040127661A1 (en) * 2002-09-12 2004-07-01 Harald Kaspar Fluoroelastomers having low temperature characteristics and solvent resistance
US20040115727A1 (en) * 2002-12-11 2004-06-17 Allergan, Inc., A Corporation Evolved clostridial toxins with altered protease specificity
US20040151741A1 (en) * 2002-12-20 2004-08-05 Gary Borodic Pharmaceutical botulinum toxin compositions
US20060165657A1 (en) * 2003-02-05 2006-07-27 Paul Bernasconi Method for delivery of cosmetic by topical application
US20060153876A1 (en) * 2003-02-24 2006-07-13 Ira Sanders Cell membrane translocation of regulated snare inhibitors, compositions therefor, and methods for treatment of disease
US20040229038A1 (en) * 2003-03-03 2004-11-18 Elan Pharma International Ltd. Nanoparticulate meloxicam formulations
US20050208083A1 (en) * 2003-06-04 2005-09-22 Nanobio Corporation Compositions for inactivating pathogenic microorganisms, methods of making the compositons, and methods of use thereof
US20060153877A1 (en) * 2003-06-20 2006-07-13 Shunji Kozaki Remedies for dissease with hypermyotonia
US20050079131A1 (en) * 2003-08-08 2005-04-14 Lanza Gregory M. Emulsion particles for imaging and therapy and methods of use thereof
US20050048088A1 (en) * 2003-08-28 2005-03-03 Fred Zulli Compositions comprising at least two nanoemulsions
US20050096340A1 (en) * 2003-10-29 2005-05-05 Yuehua Zhang Tocopherol-modified therapeutic drug compounds
US20050184275A1 (en) * 2004-02-23 2005-08-25 The Texas A&M University System Antioxidant compositions and methods of use thereof
US20050196414A1 (en) * 2004-03-03 2005-09-08 Essentia Biosystems, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US20050214325A1 (en) * 2004-03-26 2005-09-29 Vvii Newco 2003, Inc. Compositions and methods to increase the effect of a neurotoxin treatment
US20050249686A1 (en) * 2004-04-27 2005-11-10 Francoise Pataut Cosmetic composition free of water comprising fatty acid esters, petrolatum oil and non-ionic polymers
US7228259B2 (en) * 2004-06-30 2007-06-05 Lucent Technologies Inc. Method and apparatus for structure-preserving reduced-order modeling
US20060018931A1 (en) * 2004-07-26 2006-01-26 Taylor Harold V Therapeutic composition with a botulinum neurotoxin
US20060057165A1 (en) * 2004-09-10 2006-03-16 Dimitrios Dimitrakoudis Clostridium botulinum toxin formulation and method for reducing weight
US20060073208A1 (en) * 2004-10-01 2006-04-06 Allergan, Inc. Cosmetic neurotoxin compositions and methods
US20070036831A1 (en) * 2005-08-09 2007-02-15 Nanobio Corporation Nanoemulsion compositions having anti-inflammatory activity
US20070148194A1 (en) * 2005-11-29 2007-06-28 Amiji Mansoor M Novel nanoemulsion formulations
US20070178121A1 (en) * 2006-01-27 2007-08-02 Allergan, Inc. Methods for enhancing skin treatments
US20100183726A1 (en) * 2006-08-02 2010-07-22 Robert Nicolosi Compositions and methods for treating cancer with dacarbazine nanoemulsions
US20110206736A1 (en) * 2008-09-23 2011-08-25 Thomas Jefferson University Cancer Vaccines Against Mucosal Antigens and Methods of Making and Using the Same

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10016364B2 (en) 2005-07-18 2018-07-10 University Of Massachusetts Lowell Compositions and methods for making and using nanoemulsions
US20090306198A1 (en) * 2005-09-16 2009-12-10 Robert Nicolosi Anti-Oxidant Synergy Formulation Nanoemulsions to Treat Caner
US8318181B2 (en) 2005-12-01 2012-11-27 University Of Massachusetts Lowell Botulinum nanoemulsions
US10576034B2 (en) 2005-12-01 2020-03-03 University Of Massachusetts Lowell Botulinum nanoemulsions
US10532019B2 (en) 2005-12-01 2020-01-14 University Of Massachusetts Lowell Botulinum nanoemulsions
US9486408B2 (en) 2005-12-01 2016-11-08 University Of Massachusetts Lowell Botulinum nanoemulsions
US20100183726A1 (en) * 2006-08-02 2010-07-22 Robert Nicolosi Compositions and methods for treating cancer with dacarbazine nanoemulsions
US20150030671A1 (en) * 2006-11-27 2015-01-29 Zomanex, Llc Methods and formulations for enhancing the absorption and decreasing the absorption variability of orally administered drugs, vitamins and nutrients
US20080124387A1 (en) * 2006-11-27 2008-05-29 Kapac, Llc Methods and formulations for enhancing the absorption and decreasing the absorption variability of orally administered drugs, vitamins and nutrients
US20110111009A1 (en) * 2006-11-27 2011-05-12 Zomanex Llc Methods and formulations for enhancing the absorption and decreasing the absorption variability of orally administered drugs, vitamins and nutrients
US10758485B2 (en) 2006-12-01 2020-09-01 Anterios, Inc. Amphiphilic entity nanoparticles
US9724299B2 (en) 2006-12-01 2017-08-08 Anterios, Inc. Amphiphilic entity nanoparticles
US10905637B2 (en) 2006-12-01 2021-02-02 Anterios, Inc. Peptide nanoparticles and uses therefor
US10285941B2 (en) 2006-12-01 2019-05-14 Anterios, Inc. Amphiphilic entity nanoparticles
US9486409B2 (en) 2006-12-01 2016-11-08 Anterios, Inc. Peptide nanoparticles and uses therefor
US10016451B2 (en) 2007-05-31 2018-07-10 Anterios, Inc. Nucleic acid nanoparticles and uses therefor
US20100330135A1 (en) * 2007-07-06 2010-12-30 Fujifilm Corporation Cosmetic composition
US20100197785A1 (en) * 2007-07-25 2010-08-05 Epax As Omega-3 fatty acid fortified composition
US20110206739A1 (en) * 2008-03-28 2011-08-25 University Of Massachusetts Compositions and methods for the preparation of nanoemulsions
US8722131B2 (en) 2010-09-07 2014-05-13 Dsm Nutritional Products Ag Comestible emulsions
US20150342898A1 (en) * 2012-12-21 2015-12-03 Advanced Medical Frontier Co., Ltd. Composition and food or drink
US11452300B2 (en) 2013-03-15 2022-09-27 Leading Edge Innovations, LLC Compositions having an oil-in-water dispersion of submicron particles to enhance foods and beverages
US20140271877A1 (en) * 2013-03-15 2014-09-18 Leading Edge Innovations, LLC Substantially surfactant-free, submicron dispersions of hydrophobic agents containing high levels of water miscible solvent
US9980886B2 (en) * 2013-03-15 2018-05-29 Leading Edge Innovations, LLC Substantially surfactant-free, submicron dispersions of hydrophobic agents containing high levels of water miscible solvent
US20160250111A1 (en) * 2013-03-15 2016-09-01 Leading Edge Innovations, LLC Substantially surfactant-free, submicron dispersions of hydrophobic agents containing high levels of water miscible solvent
US9357770B2 (en) * 2013-03-15 2016-06-07 Leading Edge Innovations, LLC Substantially surfactant-free, submicron dispersions of hydrophobic agents containing high levels of water miscible solvent
US10531674B2 (en) 2013-03-15 2020-01-14 Leading Edge Innovations, LLC Compositions having an oil-in-water dispersion of submicron particles to enhance foods and beverages
US9308154B2 (en) * 2013-10-09 2016-04-12 Greenjoy Biotech Co., Ltd. Method for producing elastic vesicles
US20150098986A1 (en) * 2013-10-09 2015-04-09 Sky Tree and Camellias Co., Ltd. Method for Producing Elastic Vesicles
US20170209372A1 (en) * 2014-08-01 2017-07-27 Hovione International Ltd A Method of Preparing Amorphous Solid Dispersion in Submicron Range by Co-Precipitation
US20160089320A1 (en) * 2014-09-29 2016-03-31 Barrie Tan Non-Synthetic Emulsion-Based Lipid Formulations and Methods of Use
US10596117B1 (en) * 2014-12-31 2020-03-24 Eric Morrison Lipoleosomes as carriers for aromatic amide anesthetic compounds
US20190110986A1 (en) * 2015-10-08 2019-04-18 Nof Corporation O/w type emulsion
US11395798B2 (en) * 2015-10-08 2022-07-26 Nof Corporation O/W type emulsion
US10772345B2 (en) 2015-11-25 2020-09-15 Pepsico, Inc. Beverage nanoemulstions produced by high shear processing
WO2017091462A1 (en) * 2015-11-25 2017-06-01 Pepsico, Inc. Beverage nanoemulstions produced by high shear processing
US11311496B2 (en) 2016-11-21 2022-04-26 Eirion Therapeutics, Inc. Transdermal delivery of large agents
US10206417B1 (en) * 2018-06-07 2019-02-19 King Saud University Guava seed (Psidium guajava) nanoparticles as antibacterial agent
US11465107B2 (en) * 2019-01-31 2022-10-11 Hong Ngoc Thi Dang Process for producing a nano omega-3 microemulsion system
US11622947B2 (en) 2019-05-31 2023-04-11 American River Nutrition, Llc Compositions comprising quillaja extract and methods of preparations and use thereof
US20220054414A1 (en) * 2020-08-19 2022-02-24 Readymix Foods Corp. Nanoemulsion Compositions Comprising Saponins for Increasing Bioavailability
CN112042928A (en) * 2020-08-31 2020-12-08 华南理工大学 Method for synergistically and efficiently preparing protein-based nano emulsion by using polyhydroxy alcohol as molecular chaperone and prepared protein-based nano emulsion

Also Published As

Publication number Publication date
AU2006346369A1 (en) 2008-01-24
IL243260A0 (en) 2016-02-29
KR101430767B1 (en) 2014-08-19
EP1919447A2 (en) 2008-05-14
CA2618655A1 (en) 2008-01-24
BRPI0613631A2 (en) 2011-01-18
CN101384247A (en) 2009-03-11
US10016364B2 (en) 2018-07-10
BRPI0613631A8 (en) 2017-12-26
CA2618655C (en) 2015-12-22
US20150335574A1 (en) 2015-11-26
KR20130079645A (en) 2013-07-10
IL188666A0 (en) 2011-08-01
WO2008010788A3 (en) 2008-10-23
CN101384247B (en) 2013-05-22
WO2008010788A9 (en) 2008-03-06
CN103315954A (en) 2013-09-25
KR20080058326A (en) 2008-06-25
AU2006346369B2 (en) 2013-02-21
JP2009501802A (en) 2009-01-22
JP5368793B2 (en) 2013-12-18
IL188666A (en) 2016-03-31
WO2008010788A2 (en) 2008-01-24
EP1919447A4 (en) 2012-10-31
JP2013028615A (en) 2013-02-07

Similar Documents

Publication Publication Date Title
US10016364B2 (en) Compositions and methods for making and using nanoemulsions
US11260033B2 (en) Compositions for the delivery of therapeutic agents and methods of use and making thereof
TWI290052B (en) Emulsion vehicle for poorly soluble drugs
WO2007035311A2 (en) Anti-oxidant synergy formulation nanoemulsions to treat cancer
Xue et al. Anti-aging properties of phytoconstituents and phyto-nanoemulsions and their application in managing aging-related diseases
US11173127B2 (en) Potato protein nanoparticles
AU2018220054B2 (en) Compositions and methods for making and using nanoemulsions
AU2013200590B2 (en) Compositions and methods for making and using nanoemulsions
US20220054414A1 (en) Nanoemulsion Compositions Comprising Saponins for Increasing Bioavailability
Kaur Nanoemulsions as delivery vehicles for food and pharmaceuticals
Raktate et al. Encapsulation of functional foods in nanoemulsion
Sridhar et al. Recent Advances on Nanoparticle Based Strategies for Improving Carotenoid Stability and Biological Activity. Antioxidants 2021, 10, 713
Leibtag A Comprehensive Study on Ultrasonically Prepared Nanoemulsions: Formulation Development, Influence of Processing Parameters and Efficacy as Hydrophobic Drug Carriers
WO2023283277A1 (en) Fermented uni-sourced nanoemulsion of nigella sativa or cannabis sativa for use in medical, cosmetic, and recreational indications with a method of its production and use
Bhatt et al. Nanoemulsion through cold emulsification: An advanced cold manufacturing process for a stable and advanced drug delivery system

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF MASSACHUSETTS LOWELL, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NICOLOSI, ROBERT;WILSON, THOMAS A;REEL/FRAME:021225/0057;SIGNING DATES FROM 20080208 TO 20080228

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION