WO1997004747A1 - Drug delivery systems for macromolecular drugs - Google Patents

Drug delivery systems for macromolecular drugs Download PDF

Info

Publication number
WO1997004747A1
WO1997004747A1 PCT/US1996/012203 US9612203W WO9704747A1 WO 1997004747 A1 WO1997004747 A1 WO 1997004747A1 US 9612203 W US9612203 W US 9612203W WO 9704747 A1 WO9704747 A1 WO 9704747A1
Authority
WO
WIPO (PCT)
Prior art keywords
controlled release
pharmaceutical formulation
release pharmaceutical
poly
acid
Prior art date
Application number
PCT/US1996/012203
Other languages
French (fr)
Inventor
James M. Dunn
Original Assignee
Dunn James M
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 Dunn James M filed Critical Dunn James M
Publication of WO1997004747A1 publication Critical patent/WO1997004747A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5161Polysaccharides, e.g. alginate, chitosan, cellulose derivatives; Cyclodextrin

Definitions

  • the present invention relates to improved pharmaceutical formulations for administration of macromolecules or drugs that are not bioavailable using standard pharmaceutical methods. More specifically, the invention relates to pharmaceutical formulations of controlled release rate dosages of the drugs that may be administered orally, parenterally, or by inhalation.
  • Oral delivery of drugs or long acting parenteral dosage forms of drugs is preferred by the patient and physician because of compliance and the inherent beneficial effect of constant pharmacodynamic action.
  • a drug to be absorbed by the intestine it must first become soluble in the aqueous media of the gut. Products that are rapidly dissolved in water are usually rapidly absorbed into the body.
  • these drugs when given parenterally may have short half lives or may not have effective injection delivery systems. This problem arises from the fact that the drugs are destroyed before being absorbed or they are of such a size and nature that the body cannot absorb the medication. Similarly, their injectable forms have short durations of action, requiring frequent injections, making the products unsuitable for use in the non hospitalized patient.
  • the driving force for passive drug diffusion is the difference between the concentration gradient of the diffusing drug in the intestinal tract and the concentration gradient on the other side of the plasma membrane.
  • the rate of drug penetration-diffusion corresponds to the concentration gradient as is characterized by
  • the present invention obviates the problems of solvation as well as the need for classic abso ⁇ tion for delivery of those drugs where the molecular size is too large to be absorbed when the product is formulated and administered, or because of the deleterious effects of gastrointestinal enzymes, and other factors.
  • the invention is directed to pharmaceutical formulations for effective controlled release of many drugs not now orally or parenterally available. These formulations, properly adjusted, also may be administered by inhalation and achieve the same kinetic characteristics.
  • the pharmaceutical formulations of this invention are made by entrapping the drug of choice in either an organic or water phase biodegradable hydrogel polymer system to produce nanoparticles.
  • the advantage of the nanoparticle system is that its absorption by the body is by the lymphatic or lacteal system. Because of the resistant hydrogel coating it is not affected by the enzymes or degradative influences present in the gastrointestinal tract. Using bioadhesive hydrogel polymers ensures a more prolonged duration of time in which the nanoparticles are in contact with the intestinal mucosa, obfuscating the deleterious action of heightened gastrointestinal peristalsis. All of these compounds work in harmony to produce viable products that have demonstrated favorable and reproducible effects in animals.
  • the invention thus describes simple and predictable methods for the preparation of oral, parenteral, or inhalation dosage forms of a drug or drugs entrapped in biodegradable hydrogel polymers.
  • the following combinations may be used to formulate a controlled release pharmaceutical product:
  • a controlled release pharmaceutical formulation with a biologically active molecule which is encapsulated with a cyclodextrin, entrapped with liposomes and formed into nanoparticles then with water-soluble hydrogel polymers, and then coated with one or more bioadhesive adjuvants.
  • the sphere size in the nanoparticle will typically be in the range of 500 to 1500 nm, so that the drug(s) so entrapped may be administered orally, parenterally, or by inhalation.
  • the exact diameter of the nanoparticles is not critical, provided that is sufficiently small for cell diffusion by the lymphatic or lacteal system.
  • the rate of active drug release from the entrapped spheres is dependent upon the basic kinetics of the drug administered, the amount of hydrogel polymer, the cyclodextrin used, the phospholipid coating and, in the case of parenteral administration, the method of administration, the area of deposition, and the vascularity of the body region.
  • inhalation dosing it is not necessary to use the bioadhesive adjuvants, although the adjuvants may be included in these formulations.
  • Heparin is a well-established drug that is a complex polysaccharide that is highly charged (electronegative). It has a molecular weight of 20,000-60,000 daltons, which are covalently attached to a core protein found in most secretory cells. Heparin is a multifaceted drug with primary actions as an anticoagulant. When venous or arterial thrombi occur and the patient survives, heparin is given at doses of 10,000-20,000 units every 4-6 hours to maintain blood levels at 0.5-1.5 anti-factor Xa units/mL, which should prevent further thromboembolic phenomena. After this, the heparin dose is usually maintained at 10,000 units every 4-6 hours to prevent further thromboembolic events.
  • heparin has the ability to bind to the arterial wall following angioplasty and ameliorate the proliferation of smooth muscle cells and ameliorate the restenosis that so often occurs with this procedure. Also, heparin is used as a preventive agent for those patients that are at close risk of stroke or heart attack as well as the patients recovering from a heart attack.
  • Heparin also has a clearing effect in the blood by activating lipoprotein lipase on the cell surface. This action clears hyperlipoproteinmia and lowers the low density lipoprotein. In animals, it has reversed the athrogenic deposits on the arterial walls, which is a phenomenon of arteriosclerosis in humans.
  • Insulin is a polypeptide with two peptide chains linked by a disulfide bridge.
  • the A chain of insulin contains 21 amino acids and the B peptide chain contains 30 amino acids. This structural integrity is important for its biologic activity. If the amino acid sequences are disrupted or the disulfide bridge broken, the hormone becomes inactivated.
  • biodegradable polymers poly (lactic acid), poly (glycolic acid), (poly (e-caprolactone), poly (p-hydrohybutyrate), poly (b-hydroxyvalerate).
  • methacrylate polymers can be used specifically in water soluble systems. These include, but are not limited to acrylic acid, methacrylacetic cyanoethyl methocrylitic aminoakyl methacrylate copolymers, ethoxyethyl methacrylate copolymers, polymethacrylic acid, methacrylate acid alkylamide copolymer, polymethacrylic acid, polyacrylic acid, methacryic acid alkylamide copolymer, polymethacrylic acid (anhydride), and polymethyl methacrylate.
  • the advantage of the acrylate series is that when properly compounded they are water soluble or can be supplied as a 30% aqueous solution with varying degrees of solubility.
  • polymers known to the art can be used in either the water or organic phase, if properly prepared.
  • An example of this is alginic acid in intimate admixture with the drug, then cross-linking the alginic acid with a known inorganic element such as zinc, magnesium, calcium, or sodium salts.
  • pectin, zein, and guar gum can be used and cross-linked with a metallic ion.
  • Ethyl cellulose can also be used as a polymer to entrap the molecule and can be used in an aqueous or organic solvent system.
  • Cyclodextrins are a new class of compounds derived from corn. They are cyclic, non-reducing oligosaccharides built up from six, seven, and eight glucopyranose rings known respectively as alpha, beta, and gamma cyclodextrins. In addition, hydroxypropyl and other groups have been and are added to the molecule giving each series its own specific characteristics and pharmacological behavior. These molecules have a unique shape as shown in Figure 1.
  • the cyclodextrins act as a biodegradable trapping agent, inco ⁇ orating the active medication into its core and forming a unique molecular inclusion complex. These complexes provide an anchoring compound with the formation of covalent bonds. This guest-host complex protects the active drug, while not forming tight chemical bonds.
  • Cyclodextrins can be used to entrap the drug, providing a guest-host interaction.
  • Cyclodextrins are usually used in amounts that range from 0.25%-2% W/W, depending upon the bioactive molecule and the degree of protection required. The type of cyclodextrins used will again depend upon the molecule and the degree of protection required. These parameters can easily be determined by routine experimentation by one of skill in the art.
  • Liposomes were first characterized in 1965. They are single or combinations of lipids formed into spherical shapes with drugs or other active medicaments. Lipids or liposomes, primarily phospholipids, are, in general, readily absorbed intact by the intestinal cells, degraded as they pass through the interstitial space and slowly reconstituted by the body where they are then excreted by active transport in the ileum and colon in a newly reconfigured formulation. About 30-40% of the fecal dry weight is fat or lipid. Most importantly, essentially all the fat or lipids ingested are absorbed directly into the portal blood and lymphatic system or by transcell migration. Liposomes, like their parent lipids, can enter the system in a number of ways:
  • Intermembrane transfer which occurs when liposomes are in close contact with the cell they are exchanged with certain lipids in the cell wall, where they are transported intact into the cell.
  • Fusion which occurs when there is a close approach of the lipids in the cell wall and those of the liposomal lipids. Fusion results in complete mixing of the plasma membrane and release of the liposomal contents into the cytoplasm of the cell.
  • liposomes have been used for some drug delivery, there are problems with manufacturing, stability, and assurance of release. Their advantage is that drugs encapsulated into a lipid sphere can carry an electrostatic charge, allowing them to interact with cells of the opposite charge. By such a process of fusion and constant release, they efficiently can deliver the active drug moiety to the body.
  • the preferred agents are cholesterol, lecithin, glycerol monostearate, phospholipon 90H (Natterman) maleated soybean oil, sphingomylein, phosphatidyl entanolamine, dipamitoyl phoisphophatidykaline, phophosinositol, phophatidic acid, triglycerides. Since lipids tend to undergo oxidative degeneration, alpha tocopherol is usually added as an ancillary agent along with other antioxidants when the drug and or the liposomes.
  • the preferred bioadhesive adjuvant(s) that is added only to the total formulation are hydroxypropyl methyl cellulose, methyl cellulose, pectin, guar gum, xantham gums, gum acacia, gum dragon, hydroxypropyl alginate, sodium carboxymethyl cellulose, carbomer 934-P, acrylic acid derivatives and those of similar pharmaceutical characteristics and behavior.
  • These adjuvants are pharmacopeial items and are blended in with the nanoparticle granulate at the final stage of production.
  • the preferred dose is a single or combination of these adjuvants at a 50/50 W/W ratio and a percentage weight of the total granulate of 0.1-3% preferably in the range of 0.3-1.5% and most preferably 0.2-1.2 weight percentage.
  • the invention describes the formulation of a range of products with a variety of process steps and combinations. Embodiments in addition to those illustrated will be readily understood by those skilled in the art.
  • the present invention includes within its scope novel processes and products derived from the invention whether as individual features or in combination with each other to produce novel combinations.
  • the invention is illustrated further by the following examples, which are not to be taken as limiting in any way.
  • gastrointestinal absorption of oral formulations of water soluble dyes entrapped into nanoparticles, prepared according to the present invention, were studied by light microscopy.
  • Nanoparticles containing fluorescent stains were prepared and administered orally with concomitant bioadhesive adjuvants, as a single dose to anesthetized rabbits, via a gastric tube.
  • the rabbits were sacrificed 7 and 14 days after oral administration of the manufactured spheres. Both ultraviolet light microscopy and direct vision revealed dye-containing spheres widely distributed throughout the animal's bodies.
  • the yield was 742 mg out of a theoretical yield of 1,400 mg.
  • the resultant powder/granulate was weighed and titrated against protamine in normal saline to ensure that heparin was in fact entrapped in the spheres. To do this, the beads were suspended in normal saline and agitated by vortex. Protamine was then added to the solution and no precipitate was observed. The nanoparticles were assayed before l o degradation and found to have 0.75 U/ mg of heparin.
  • Electron microscopy demonstrated an average diameter of 850 nm, with a range of 650 to 1250 nm.
  • nanoparticles were coated with bioadhesive adjuvant. Nanoparticles were coated by dispersion in 20-mL of an aqueous
  • Vancomycin hydrochloride (3,000 mg) was added to 300 mL of KH2PO4 at pH 6.8
  • Heparin nanoparticles prepared according to Example 1, were orally administered to rabbits in an animal study. These preliminary results showed that heparin, a large molecular weight drug, can be formulated for oral absorption in a controlled, sustained-release fashion.
  • the rabbits were sedated with xyalazine for administration of the drugs and control.
  • a soft #10 rubber French catheter was then placed into the animals' stomach and the material was delivered by pressure to simulate a controlled oral administration.
  • the tube was rinsed with deionized water to make sure all the particles were expelled from the tube into the rabbits' stomach. In all, this process was repeated five times over a six month period with three different animals treated each time, for a total of 15 animals.
  • the animals were housed and fed rabbit chow and were given water ad lib. Blood samples were taken from the marginal ear vein at the following times, 6 hours post dose, 24, 48, 96, 120 and 144 hours after drug administration. The blood samples were placed in a citrate Eppendorf test tube immediately after blood was obtained and spun down using 3,000 Xg for 23 minutes. All samples were then assayed for antifactor Xa by Dr. Richard Malar at the University Of Colorado-Veterans Administration Hospital coagulation lab. The test was blinded as to the specimen, its timing and the specific animal. Following the centrifugation, the plasma was removed and frozen immediately at -20"C until assay.
  • thromboplastin 100 mL/mouse
  • a blood clotting activating agent a blood clotting activating agent
  • the model measures the level of protection given by the various drugs.
  • the intravenous injection of thromboplastin with the dose of 100 ⁇ L/mice (0.2 ng) provokes a thromboembolism involving 80% mortality in 24 hours.
  • the intravenous heparin standard protects 50% of animals treated when administered in the range of 0.09 mg/kg.
  • the heparin nanoparticles were given by oral route in suspension in a carboxymethyl cellulose solution (0.06%) in doses of 50, 150, and 300 mg/kg.
  • heparin nanoparticles In addition to the three dosage levels of the heparin nanoparticles tested, a blank and two heparin standards were run.
  • the blank was the bioadhesive adjuvant, carboxymethyl cellulose, at 0.6%.
  • the first heparin standard was intravenous heparin at 0.3 mg/kg.
  • the second heparin standard was 85 mg/kg administered orally.
  • the fluid was then placed on a preheated stirring plate and at very low temperature ⁇ 40°C and stirred 1700 Rpm's for 30 minutes. After stirring was complete, the fluid was placed into a modified Pyrex® dish and allowed to air dry for 24 hours. At the end of this time there was a very fine granulated powder that was removed by a sharp razor blade edge and weighed. The theoretical yield was 2.80 grams, while the actual yield was 2.75 grams.

Abstract

This invention allows for the oral, parenteral or inhalation delivery of large macromolecules. Biologically active drugs entrapped into biodegradable hydrogel polymers in either organic and water phase systems. By using cyclodextrins, sensitive molecules can be protected during the granulations of nanoparticles production phase.

Description

DRUG DELIVERY SYSTEMS FOR MACROMOLECULAR DRUGS
FIELD OF INVENTION
The present invention relates to improved pharmaceutical formulations for administration of macromolecules or drugs that are not bioavailable using standard pharmaceutical methods. More specifically, the invention relates to pharmaceutical formulations of controlled release rate dosages of the drugs that may be administered orally, parenterally, or by inhalation.
BACKGROUND OF THE INVENTION
Oral delivery of drugs or long acting parenteral dosage forms of drugs is preferred by the patient and physician because of compliance and the inherent beneficial effect of constant pharmacodynamic action. Fundamentally, for a drug to be absorbed by the intestine it must first become soluble in the aqueous media of the gut. Products that are rapidly dissolved in water are usually rapidly absorbed into the body.
For such products, controlling the rate of solvation or rate of absoφtion after ingestion also influence their bioavailability and subsequently their pharmacodynamic effectiveness.
Proteins and polypeptides, as well as some complex carbohydrates, however, are degraded by enzyme systems in the intestinal tract and are thus not orally active.
Similarly these drugs when given parenterally may have short half lives or may not have effective injection delivery systems. This problem arises from the fact that the drugs are destroyed before being absorbed or they are of such a size and nature that the body cannot absorb the medication. Similarly, their injectable forms have short durations of action, requiring frequent injections, making the products unsuitable for use in the non hospitalized patient.
It is medically desirable to deliver medicinal agents to the body in a smooth and controlled fashion. This avoids the "peaks and valleys" of most immediate release oral and parenteral preparations. Common factors influencing drug absorption is the passage of drugs across the intestinal membrane. A drug almost always has to cross several cellular membranes to reach its receptor site. This transfer across cell membranes is normally accomplished by passive diffusion. Special transport mechanisms such as facilitated diffusion and active transport allow some substances to cross cell membranes at a faster rate than simple diffusion. By far the most common procedure for drug transport across the cell membrane, however, is passive diffusion. Passive diffusion is characterized by the movement of a drug molecule down an electromagnetic or concentration gradient without the expenditure of cellular energy. The transfer process is neither saturable nor inhibited by other materials and is only slightly sensitive to temperature changes. Since most cells in the gastrointestinal tract are in proximity to capillaries, the passage of drugs across this short distance is usual rapid.
The driving force for passive drug diffusion is the difference between the concentration gradient of the diffusing drug in the intestinal tract and the concentration gradient on the other side of the plasma membrane. The rate of drug penetration-diffusion corresponds to the concentration gradient as is characterized by
Fick's law.
Many drugs are either an organic acid or a base. Acids donate a hydrogen to form a negatively charged anion while bases accept a hydrogen ion to form a positively charged cation. It is usually assumed that only nonionized, lipid-soluble drugs pass through the lipid rich membranes of the intestinal tract. The ionized molecules are thought to be too polar to penetrate this lipoid barrier. If the ionized molecule does cross the cell wall, however, it does so at a slow rate. This concept of drug absorption is known as nonionic diffusion.
An extension of this principle is the pH partition hypothesis, which states that the passage rate of a drug through a membrane is dependent upon the pH of the drug's environment and the dissociation constant, or pKa of the drug. The pKa is expressed as that pH at which 50% of the drug will be ionized and 50% will be in the nonionized form. Diffusion of acids and bases across the cell membrane is not always influenced by pH, as in the case of weak acids or bases. These types of products are essentially completely nonionized at all physiological pH values. At the other extreme, however, are strong acids and bases that are almost completely ionized, and their transfer is dependent upon the pH at which they become dissolved and subsequently become ionized or nonionized.
An example of this pH partition hypothesis may be explained by the fact that aspirin, which has a pKa, or dissociation constant of between 3-3.7, becomes very nonionized in the acid media of the stomach and subsequently is rapidly absorbed from the gastric mucosa, where the pH is between 1-3. As the drug particles pass into the small intestine where the pH increases, the rate of ionization is changed so absorption is subsequently slowed. Conversely, strong bases such as ephedrine, which has a pH of 9.3, are almost negligibly absorbed in the acid media of the gastric juice and are absorbed rather rapidly from the intestinal media which has a much higher pH.
While the pH partition hypothesis is but one of the factors influencing drug absoφtion, it cannot explain all the phenomena observed with different delivery systems and the rate of drug delivery in such systems.
SUMMARY OF THE INVENTION
The present invention obviates the problems of solvation as well as the need for classic absoφtion for delivery of those drugs where the molecular size is too large to be absorbed when the product is formulated and administered, or because of the deleterious effects of gastrointestinal enzymes, and other factors. The invention is directed to pharmaceutical formulations for effective controlled release of many drugs not now orally or parenterally available. These formulations, properly adjusted, also may be administered by inhalation and achieve the same kinetic characteristics. The pharmaceutical formulations of this invention are made by entrapping the drug of choice in either an organic or water phase biodegradable hydrogel polymer system to produce nanoparticles. These nanoparticles are then coated or combined with one or more bioadhesive adjuvants to promote adherence of the particles and their medications to the intestinal wall. There are two preferred pharmaceutical formulation embodiments to form the nanoparticles: In the first instance, drugs that are rugged and can withstand organic solvents are treated by entrapment in single or combinations of biodegradable hydrogel polymers. Alternatively, non-organic solvents may be used, but it is preferred to entrap drugs that need a water-based system in a cyclodextrin. This entrapped drug nanoparticle complex may then secondarily be entrapped in liposomes.
With those drugs that are delicate to changes in their environment, the drugs can first be entrapped into a cyclodextrin for protection, then granulated with water soluble, hydrogel polymers. These entrapped cyclodextrin-drug complexes can be secondarily entrapped in liposomes before or after their enclosure into the hydrogel. The invention is therefore suitable for using both organic and non-organic solvents.
The advantage of the nanoparticle system is that its absorption by the body is by the lymphatic or lacteal system. Because of the resistant hydrogel coating it is not affected by the enzymes or degradative influences present in the gastrointestinal tract. Using bioadhesive hydrogel polymers ensures a more prolonged duration of time in which the nanoparticles are in contact with the intestinal mucosa, obfuscating the deleterious action of heightened gastrointestinal peristalsis. All of these compounds work in harmony to produce viable products that have demonstrated favorable and reproducible effects in animals.
The invention thus describes simple and predictable methods for the preparation of oral, parenteral, or inhalation dosage forms of a drug or drugs entrapped in biodegradable hydrogel polymers. The following combinations may be used to formulate a controlled release pharmaceutical product:
A controlled release pharmaceutical formulation with a biologically active molecule which is formed into nanoparticles with biodegradable hydrogel polymers and then coated with one or more bioadhesive adjuvants.
A controlled release pharmaceutical formulation with a biologically active molecule which is formed into nanoparticles with biodegradable hydrogel polymers, coated with liposomes, and then coated with one or more bioadhesive adjuvants. A controlled release pharmaceutical formulation with a biologically active molecule encapsulated with a cyclodextrin, which is then formed into nanoparticles with biodegradable water-soluble hydrogel polymers, and then coated with one or more bioadhesive adjuvants.
A controlled release pharmaceutical formulation with a biologically active molecule which is encapsulated with a cyclodextrin, entrapped with liposomes and formed into nanoparticles then with water-soluble hydrogel polymers, and then coated with one or more bioadhesive adjuvants.
A controlled release pharmaceutical formulation with a biologically active molecule encapsulated with a cyclodextrin that is then formed into nanoparticles with biodegradable water-soluble hydrogel polymers, coated with liposomes, and then coated with one or more bioadhesive adjuvants.
The sphere size in the nanoparticle will typically be in the range of 500 to 1500 nm, so that the drug(s) so entrapped may be administered orally, parenterally, or by inhalation. The exact diameter of the nanoparticles is not critical, provided that is sufficiently small for cell diffusion by the lymphatic or lacteal system.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows the chemical nature of the cyclodextrin skeleton. Figure 2A is a photomicrograph of a scanning electron microscope at 30,000X magnification, showing size and diameter of heparin nanoparticles. Figure 2B is a photomicrograph of a scanning electron microscope at 6,000X demonstrating clusters of nanoparticles.
Figure 3 is a graphic display comparing the levels of plasma heparin from Table 1. This figure shows the mean (+SD) plasma heparin values for 12 rabbits given oral heparin nanoparticles plus adjuvant (2,400 U/kg), and for 1 rabbit each given oral heparin (11,000 U/kg), oral heparin (11,000 U/kg) plus adjuvant, or adjuvant alone.
DETAILED DESCRIPTION OF THE INVENTION
Simple and predictable methods have been discovered for the preparation of controlled release active dosage formulations of drugs entrapped in biodegradable hydrogel polymers, with alternative concomitant entrapment in a protective quantity of cyclodextrins. The polymer/drug complex and the polymer/drug/cyclodextrin complex may then be coated with phospholipids or other lipoid components forming secondary liposomes. These combinations are then combined with an appropriate quantity of bioadhesive adjuvant. The drugs so entrapped may be administered orally, parenterally, or by inhalation.
The rate of active drug release from the entrapped spheres is dependent upon the basic kinetics of the drug administered, the amount of hydrogel polymer, the cyclodextrin used, the phospholipid coating and, in the case of parenteral administration, the method of administration, the area of deposition, and the vascularity of the body region. With inhalation dosing, it is not necessary to use the bioadhesive adjuvants, although the adjuvants may be included in these formulations.
The invention is thus directed to controlled release rate formulations of biologically active molecules, especially macromolecules. Any therapeutic drug, biological active protein or polypeptide, polysaccharide molecule, carbohydrate complex vaccine, such as an influenza vaccine, and the like, referred to generally herein as "bioactive material," "biologically active molecule," or "drug," can be formulated according to this invention. In addition, more than one drug can be formulated together according to this invention. Primary candidates for development into oral formulations are the widely used drugs, particularly those drugs with low bioavailability and drugs currently only capable of being administered via parenteral means. Two of the preferred drugs that can be prepared according to this invention are heparin and insulin. These drugs are classic large molecular weight drugs that have been considered the gold standard for effectiveness of new drug delivery systems for large polypeptide or carbohydrate drugs, and have previously been unavailable by oral means. With the pharmaceutical formulations of this invention, these drugs are not only absorbed orally, but their pharmacological effects are prolonged.
Heparin is a well-established drug that is a complex polysaccharide that is highly charged (electronegative). It has a molecular weight of 20,000-60,000 daltons, which are covalently attached to a core protein found in most secretory cells. Heparin is a multifaceted drug with primary actions as an anticoagulant. When venous or arterial thrombi occur and the patient survives, heparin is given at doses of 10,000-20,000 units every 4-6 hours to maintain blood levels at 0.5-1.5 anti-factor Xa units/mL, which should prevent further thromboembolic phenomena. After this, the heparin dose is usually maintained at 10,000 units every 4-6 hours to prevent further thromboembolic events.
In addition, heparin has the ability to bind to the arterial wall following angioplasty and ameliorate the proliferation of smooth muscle cells and ameliorate the restenosis that so often occurs with this procedure. Also, heparin is used as a preventive agent for those patients that are at close risk of stroke or heart attack as well as the patients recovering from a heart attack.
Heparin also has a clearing effect in the blood by activating lipoprotein lipase on the cell surface. This action clears hyperlipoproteinmia and lowers the low density lipoprotein. In animals, it has reversed the athrogenic deposits on the arterial walls, which is a phenomenon of arteriosclerosis in humans.
Another of the drugs that has been previously studied for oral administration is insulin. Insulin is a polypeptide with two peptide chains linked by a disulfide bridge. The A chain of insulin contains 21 amino acids and the B peptide chain contains 30 amino acids. This structural integrity is important for its biologic activity. If the amino acid sequences are disrupted or the disulfide bridge broken, the hormone becomes inactivated.
According to the present invention, two methods are provided for incorporating bioactive molecules in nanoparticles. The first method covers materials that are soluble in organic solvents; the second method is for those molecules that are water soluble only.
BIODEGRADABLE HYDROGEL POLYMERS
Microparticles (<900 nm - 10 nm) have been used for a number of years as a drug delivery method. This is when hydrogel polymers are allowed to entrap an organic molecule, then fixated it into spheres, by several methods. This allows the drug to enter the body protected from hazardous enzymatic digestion in the gastrointestinal tract. In general, the spheres are produced by dissolving the active medicinal agent in a suitable solvent such as buffered water or purified water. The hydrogel, which are polymers of biodegradable compounds, are most often soluble in organic agents such as dichlomethane, acetone, and the like. The dispersed polymers are solvated and then added to the water-drug combination and spheres of micro particle size are formed.
These spheres, entrapped in biodegradable material, are somewhat immune to the deleterious effects of the gastrointestinal enzyme. Since they are biodegraded, they can be properly formulated so that oral and parenteral administration is possible.
Hydrolysis of the polymer breakdown is most desirable since this will produce low molecular weight by-products. Some of the more common biodegradable polymers are: poly (lactic acid), poly (glycolic acid), (poly (e-caprolactone), poly (p-hydrohybutyrate), poly (b-hydroxyvalerate). poly (tartonic acid), polydixamone, poly (ethylene terephalate) poly (ortho esters), polyanhydrides, dextran, cross-linked polyvinyl alcohol, polyhydroxy methacrylate, polycyanoacrylates, poly (phosphoesters), polyphosphgenes, poly (hydroxy butyrate co-valerate), poly (2-hydroxyethyl glutamate), polyvinylpyrrolidone, poly (ethylene glycol), poly (propylene glycol), chitosan, pullulan, zein, alginic acid and alginate salts.
In addition, other methacrylate polymers can be used specifically in water soluble systems. These include, but are not limited to acrylic acid, methacrylacetic cyanoethyl methocrylitic aminoakyl methacrylate copolymers, ethoxyethyl methacrylate copolymers, polymethacrylic acid, methacrylate acid alkylamide copolymer, polymethacrylic acid, polyacrylic acid, methacryic acid alkylamide copolymer, polymethacrylic acid (anhydride), and polymethyl methacrylate. The advantage of the acrylate series is that when properly compounded they are water soluble or can be supplied as a 30% aqueous solution with varying degrees of solubility.
Other polymers known to the art can be used in either the water or organic phase, if properly prepared. An example of this is alginic acid in intimate admixture with the drug, then cross-linking the alginic acid with a known inorganic element such as zinc, magnesium, calcium, or sodium salts. Likewise pectin, zein, and guar gum can be used and cross-linked with a metallic ion. Ethyl cellulose can also be used as a polymer to entrap the molecule and can be used in an aqueous or organic solvent system. Similar examples are polyvinyl acid phthalate, cellulose acid phthalate, cellulose acid trimaleate phthalate and similar cellulosic polymers such as hydroxypropyl methyl cellulose phthalate and the like. CYCLODEXTRINS
Cyclodextrins are a new class of compounds derived from corn. They are cyclic, non-reducing oligosaccharides built up from six, seven, and eight glucopyranose rings known respectively as alpha, beta, and gamma cyclodextrins. In addition, hydroxypropyl and other groups have been and are added to the molecule giving each series its own specific characteristics and pharmacological behavior. These molecules have a unique shape as shown in Figure 1. The cyclodextrins act as a biodegradable trapping agent, incoφorating the active medication into its core and forming a unique molecular inclusion complex. These complexes provide an anchoring compound with the formation of covalent bonds. This guest-host complex protects the active drug, while not forming tight chemical bonds.
With the drug in a buffer or suitable solution and, with agitation, cyclodextrins can be used to entrap the drug, providing a guest-host interaction. Cyclodextrins are usually used in amounts that range from 0.25%-2% W/W, depending upon the bioactive molecule and the degree of protection required. The type of cyclodextrins used will again depend upon the molecule and the degree of protection required. These parameters can easily be determined by routine experimentation by one of skill in the art.
LIPIDS AND LIPOPROTEINS Liposomes were first characterized in 1965. They are single or combinations of lipids formed into spherical shapes with drugs or other active medicaments. Lipids or liposomes, primarily phospholipids, are, in general, readily absorbed intact by the intestinal cells, degraded as they pass through the interstitial space and slowly reconstituted by the body where they are then excreted by active transport in the ileum and colon in a newly reconfigured formulation. About 30-40% of the fecal dry weight is fat or lipid. Most importantly, essentially all the fat or lipids ingested are absorbed directly into the portal blood and lymphatic system or by transcell migration. Liposomes, like their parent lipids, can enter the system in a number of ways:
1. Intermembrane transfer, which occurs when liposomes are in close contact with the cell they are exchanged with certain lipids in the cell wall, where they are transported intact into the cell.
2. Contact release, which occurs when liposomes contact the cell and there is a sudden permeability of the lipid to the interior of the cell. 3. Absorption, which occurs when there is electrochemical attraction between the cell and the liposomal particle, wherein the particle is pulled into the cell.
4. Fusion, which occurs when there is a close approach of the lipids in the cell wall and those of the liposomal lipids. Fusion results in complete mixing of the plasma membrane and release of the liposomal contents into the cytoplasm of the cell.
5. Cells with phyacytic capability take liposomes into endosomes, (which are intracellular molecules formed by imagination of the plasma membranes). Endosomes are then degraded into their component parts.
While liposomes have been used for some drug delivery, there are problems with manufacturing, stability, and assurance of release. Their advantage is that drugs encapsulated into a lipid sphere can carry an electrostatic charge, allowing them to interact with cells of the opposite charge. By such a process of fusion and constant release, they efficiently can deliver the active drug moiety to the body.
When liposomes are used as a secondary granulating agent, the preferred agents are cholesterol, lecithin, glycerol monostearate, phospholipon 90H (Natterman) maleated soybean oil, sphingomylein, phosphatidyl entanolamine, dipamitoyl phoisphophatidykaline, phophosinositol, phophatidic acid, triglycerides. Since lipids tend to undergo oxidative degeneration, alpha tocopherol is usually added as an ancillary agent along with other antioxidants when the drug and or the liposomes.
BIOADHESIVE ADJUVANTS
The preferred bioadhesive adjuvant(s) that is added only to the total formulation are hydroxypropyl methyl cellulose, methyl cellulose, pectin, guar gum, xantham gums, gum acacia, gum dragon, hydroxypropyl alginate, sodium carboxymethyl cellulose, carbomer 934-P, acrylic acid derivatives and those of similar pharmaceutical characteristics and behavior. These adjuvants are pharmacopeial items and are blended in with the nanoparticle granulate at the final stage of production. The preferred dose is a single or combination of these adjuvants at a 50/50 W/W ratio and a percentage weight of the total granulate of 0.1-3% preferably in the range of 0.3-1.5% and most preferably 0.2-1.2 weight percentage.
The invention describes the formulation of a range of products with a variety of process steps and combinations. Embodiments in addition to those illustrated will be readily understood by those skilled in the art. The present invention includes within its scope novel processes and products derived from the invention whether as individual features or in combination with each other to produce novel combinations. The invention is illustrated further by the following examples, which are not to be taken as limiting in any way.
EXAMPLES
In the examples that follow, gastrointestinal absorption of oral formulations of water soluble dyes entrapped into nanoparticles, prepared according to the present invention, were studied by light microscopy. Nanoparticles containing fluorescent stains were prepared and administered orally with concomitant bioadhesive adjuvants, as a single dose to anesthetized rabbits, via a gastric tube. The rabbits were sacrificed 7 and 14 days after oral administration of the manufactured spheres. Both ultraviolet light microscopy and direct vision revealed dye-containing spheres widely distributed throughout the animal's bodies. Microscopic observation of the tissue distribution of the dye at various times following a single oral administration of florescent containing nanoparticles suggested that absorption probably occurs by pinocytosis, as well as by direct lacteal absorption and distribution. Pinocytosis, an active transport process by which small particles of molecular or macromolecular dimensions are incorporated into intracellular vacuoles that originate by the folding of the plasma membrane around the material being engulfed, was also active.
Example 1 Formation of Heparin-Containing Nanoparticles 300 mg of heparin sodium (lmg=157 units) derived from porcine intestines, MW
~25,000 daltons, was weighed and blended in a high shear mixer with 50 mL of purified water.
To make the nanoparticles, 600 mg of Poly(3-hydroxybutyrate 3-covalerate (80:20) and 300 mg of DL-Lactic acid co-glycolide (70:30) were completely pulverized and then solvated in equal volumes of 50 mL of acetone and 50 mL of dichloromethane. The heparin and the polymers were then mixed and blended using intense high shear blending. After blending, 200 mL of deionized water was slowly added until opalescent microspheres formed. Blending was continued for an additional 10 to 15 minutes following the appearance of the microspheres, then the mixture was placed on a pre-warmed hot-plate with a magnetic stirring rod. The temperature of the plate was 60° C for 12 hours.
When the granulate was completely dried, it was scraped from the filter material and passed through a 200 mesh sieve by gentle pressure until the dried granulate was
5 homogeneous in size. The yield was 742 mg out of a theoretical yield of 1,400 mg. The resultant powder/granulate was weighed and titrated against protamine in normal saline to ensure that heparin was in fact entrapped in the spheres. To do this, the beads were suspended in normal saline and agitated by vortex. Protamine was then added to the solution and no precipitate was observed. The nanoparticles were assayed before l o degradation and found to have 0.75 U/ mg of heparin.
Acetone and methylene chloride (2 mL) were added to disrupt the beads and, upon vortex, a heavy white paste precipitate was formed. The dry granulate thus produced was then evaluated and quantified and the product observed to contain 44.3 units of heparin per milligram of granulate. The powder was also examined for is formation characteristics via a Phillips scanning electron microscope as shown in Figures
2A and 2B. Electron microscopy demonstrated an average diameter of 850 nm, with a range of 650 to 1250 nm.
To facilitate adsoφtion, the heparin-containing nanoparticles were coated with bioadhesive adjuvant. Nanoparticles were coated by dispersion in 20-mL of an aqueous
20 adjuvant containing 0.5% Carbopol-934P and 0.5% hydroxymethylcellulose.
Example 2 Water Lipid Phase Sphere Formation of Vancomycin
Vancomycin hydrochloride (3,000 mg) was added to 300 mL of KH2PO4 at pH 6.8
25 and, using a high shear mixer, was blended with 3,000 mg of chitosan and 2,000 mg of pullulan and blended at high speed for 30 minutes. After a homogeneous opalescence appeared, 300 mg of zinc chloride was added to cross link the polymers. Then 30 mL of isopropyl alcohol was added to harden the spheres. Following this, 3,000 mg of liposome phospholin 90H, a prepared phospholipid from Natterman was added and homogenized
30 for 15 minutes. The flask was then placed on a hot plate at the lowest temperature setting and magnetic stirring at 700 φm's for 48 hours. After drying, the material was placed through a vacuum and 0.1 mm filter. When dry, the cake was removed and spread on a glass plate and allowed to dry at 60°C for 24 hours. The dried material was then screened through a 200 mesh screen. The theoretical yield was 12.0 grams while the
35 actual weight after production 11.41 grams for 95.08% yield. The material was evaluated via ultraviolet spectrophotometer and a standard curve developed. The dissolution showed the vancomycin content to be 0.64 mg/mL.
Example 3 Insulin and Cyclodextrin Nanoparticles
Ten thousand units of (100 mL) aqueous insulin was admixed with beta cyclodextrin (100 gm) and blended with a Silverson high-shear mixer until a clear solution developed, indicating entrapment of the insulin in the cyclodextrin. To this solution 500 mg of liposome phospholipon 90H was added and blended, then 1000 mg as a 30% W/V dispersion of methyl methacrylate was added. The material was spread on a special "petre" Pyrex plate and allowed to dry at 60°C for 4-6 hours until dry. It was then scrapped clean with a special razor knife and was so fine that screening was unnecessary. This material was placed in a test tube for later experimentation with diabetic rats.
Example 4
Water Procedure For Entrapment In Nanoparticles Insoluble In Organic Solvents
Where the biologically active molecule to be entrapped is insoluble, or may be destroyed by organic solvents, a water phase process may be used to incorporate the agent into spheres. In this example, a red lake dye, insoluble in organic solvents, was hydrated in an aqueous solution and precipitated with organic solvents. Red lake dye
#40, 100 mg was suspended in 300 mL of ammoniated 0.05M KH2PO4 at pH 6.8 using a Silverson high shear mixer.
To this aqueous phase 3,000 mg of poly(3-hydroxybutyrate-covalerate 80:20) and poly DL-lactic acid was added and blended at high speed. Once the dye was suspended in the aqueous phase with the polymers, 1% polyvinyl alcohol W/W was added and blended with the materials as a stabilizing agent.
To this mixture was added an equal mixture of 30 mL of acetone and dichloromethane. Upon addition of the organic solvents an exothermic reaction occurred and there was the immediate formation of microparticles that were verified under light microscopy. The resultant solution was poured into 20 ml test tubes and centrifuged at
3000 g for 15 minutes. The supernatant was removed and the particulate material was repeatedly washed with deionized water until it was clear. The centrifuged material was then placed on a glass slide and allowed to air dry. After drying, there was a light pink powder that was fine and did not need screening. The granulated material was placed into deionized water and vigorously vortexed for 15 minutes. No color was apparent. 5 mL of acetone and dichloromethane were placed with the beads and again vigorously vortexed. After this treatment, a bright red color appeared. This suggested that the method as explained entrapped the dye into the nanoparticles.
Example 5
Administration Of Oral Heparin In Nanoparticles
Experiment 1 Heparin nanoparticles, prepared according to Example 1, were orally administered to rabbits in an animal study. These preliminary results showed that heparin, a large molecular weight drug, can be formulated for oral absorption in a controlled, sustained-release fashion.
Fifteen adult rabbits were randomized as follows: 12 animals received oral heparin in nanoparticles at a dose of 2,400 U/kg of body weight;
1 animal received 11,000 units of plain liquid heparin;
1 animal received liquid 11,000 units of heparin and bioadhesive adjuvant alone; and • 1 animal received only the bioadhesive adjuvant.
The rabbits were sedated with xyalazine for administration of the drugs and control. A soft #10 rubber French catheter was then placed into the animals' stomach and the material was delivered by pressure to simulate a controlled oral administration. The tube was rinsed with deionized water to make sure all the particles were expelled from the tube into the rabbits' stomach. In all, this process was repeated five times over a six month period with three different animals treated each time, for a total of 15 animals.
The animals were housed and fed rabbit chow and were given water ad lib. Blood samples were taken from the marginal ear vein at the following times, 6 hours post dose, 24, 48, 96, 120 and 144 hours after drug administration. The blood samples were placed in a citrate Eppendorf test tube immediately after blood was obtained and spun down using 3,000 Xg for 23 minutes. All samples were then assayed for antifactor Xa by Dr. Richard Malar at the University Of Colorado-Veterans Administration Hospital coagulation lab. The test was blinded as to the specimen, its timing and the specific animal. Following the centrifugation, the plasma was removed and frozen immediately at -20"C until assay. The plasma was assayed for antifactor Xa using a standard, validated chromogenic method. The heparin-containing solution (50 μL) (at various dilutions and in duplicate) was added to a microtiter well in the presence of human plasma (50 μL) and factor Xa (50 μL). The reaction was incubated for 1 minute at room temperature and 50 μL of Chromozyme Xa substrate was then added. The mixture was incubated for 10 minutes at room temperature and the reaction stopped with 50 μL of 50% acetic acid. The assay well was then measured in an enzyme-linked immunoadsorbent assay reader at 405 nm. The amount of heparin was determined based on a standard curve; the duplicate values averaged and dilutions compared. The within-run variation was +5% and the between-run variation was +9%; the lower limit of detection was 0.01 U/mL.
Values below <0.02 U/mL were considered as 0. The results are shown in Table 1 and Figure 3. The results of the three control animals show that there is no appreciable absorption of heparin under the test situations.
The results of this first experiment showed that heparin fashioned into nanoparticles produced sustained and viable plasma levels whereas regular heparin or the adjuvant given alone or together does not produce any significant levels of detectable heparin. Most importantly, this single dose produced effective doses for up to 144 hours after administration that was in stark contrast to injectable levels that seldom gave measurable levels of heparin 10-24 hours after subcutaneous injection of the drug. Table 1 is a tabular display of animal studies with rabbits showing plasma heparin values (U/mL) after oral administration of heparin nanoparticles (2400 U/kg) (test animals); heparin only (11,000 U/kg); heparin (11,000 U/kg) plus adjuvant; or adjuvant alone (control) in 15 rabbits.
TABLE 1
Anti-Factor Xa Units of Heparin Following The Administration
Of Heparin Nanospheres, Heparin, Heparin+Adjuvant, And Adjuvant
Units Of Heparin Per Milliliter U/mL
HOURS #1 #2 #3 #4 #5 #6 #7 #8 #9 #10 #11 #12 MEAN ±SD Heparin H/ADJ* co c ru 0 0 0 0 0 0 0 0 0 0 0 0 0 0.00 0.26 0 0 co 6 0.9 1.38 IJ 0.7 0.7 0.8 0.9 0.7 0.31 0.7 0.5 0.8 0.79 0.67 0.01 0.02
H 24 0.12 3.04 0.8 1 1.3 0.7 1.1 0.9 0.6 1.3 0.8 1.2 1.07 0.35 0.02 0.02 C H 48 0.25 OJ 0.9 1 1 0.8 0.9 IJ 0.35 1.2 1 IJ 0.81 0.56 0.07 0.01 m 72 0.9 0.29 IJ 0.8 0.8 2.3 1.3 1.6 0.16 0.8 1.3 0.6 1.00 0.23 0.09 0.03
0) 96 0.8 0.36 1 1 0.8 1 0.7 0.8 0.3 0.6 0.5 0.9 0.73 0.45 0.08 0.08
I m 120 0.6 0.37 2 0.9 0.9 0.8 1.2 0.5 0J7 0.6 0.7 0.7 0.79 0.21 0.L2 0.01 m 0.9 0.7 0.7 0.6 0.4 0.3 0.5 0.2
H 144 0.7 0.31 0.8 0.5 0.55 0.21 0.09 0.02 c r- m H/ADJ = Heparin + Adjuvant t ADJ = Adjuvant Only σ»
Experiment 2 Heparin nanoparticles, prepared as described in Example 1, were independently tested on an experimental mouse model that is sensitive to parenteral (injected) heparin. This experiment was performed in the research laboratories of Sanofi Pharma in France. Sanofi used a different animal species (mice) and different methods for evaluating heparin activity than that described in Experiment 1.
In this mouse model, a lethal dose of thromboplastin (100 mL/mouse), a blood clotting activating agent, was given intravenously 2 hours following various doses of drugs. The model then measures the level of protection given by the various drugs. The intravenous injection of thromboplastin with the dose of 100 μL/mice (0.2 ng) provokes a thromboembolism involving 80% mortality in 24 hours. The intravenous heparin standard protects 50% of animals treated when administered in the range of 0.09 mg/kg. In this study, the heparin nanoparticles were given by oral route in suspension in a carboxymethyl cellulose solution (0.06%) in doses of 50, 150, and 300 mg/kg. In addition to the three dosage levels of the heparin nanoparticles tested, a blank and two heparin standards were run. The blank was the bioadhesive adjuvant, carboxymethyl cellulose, at 0.6%. The first heparin standard was intravenous heparin at 0.3 mg/kg. The second heparin standard was 85 mg/kg administered orally.
The results indicated in Table 2 show that the heparin nanoparticles protected 25% of mice treated with a dose of 150 mg/kg. The heparin standard had no effect in an oral dose of 85 mg/kg. The means of determining the effect of protection was done by subtracting the negative and positive controls from the test and determining the percentage of effectiveness.
It should be noted that the heparin nanoparticles have a total duration of measurable activity for 7 days and its peak effect is from 24 to 48 hours. Nonetheless, in this study, the activity was effectively measured only 2 hours after administration of the heparin by the injection of thromboplastin. Thus, the bias in the study is against the heparin nanoparticles. Protection was still found with heparin nanoparticles at 2 hours, which showed that the heparin nanoparticles produced an immediately active burst effect. In addition, the study showed that the heparin nanoparticles increased the intestinal adsorption of heparin. Table 2
Treatment Dose Survival % Protection
Vehicle NG 2/10 20%
Heparin IV 0.3 mg/kg 8/10 80%
Heparin Nanoparticles 50 mg/kg 1/10 10%
150 mg/kg 4/10 40%
300 mg/kg 9/20 55%
Heparin liquid 85 mg/kg/po 2/10 20%
Example 6 Insulin Nanoparticles And Their Effect Upon Diabetic Rats
Since its discovery, insulin has been attempted to be given by means other than injection. The following shows the administration of insulin formulating according to this invention to animals in animal models.
Formulation
Insulin (10,000 units) as a concentrated liquid dosage was placed in a beaker and, using the Silverson mixer, blended at moderate speeds. Beta cyclodextrin was added at 1000 mg and blended with the insulin. Initially there was a milky white appearance that rather rapidly dissipated upon stirring. To this mixture was added a phospholipid at a strength of 500 mg. The agitation was continued until clearing occurred. At this point Eudragit® 30 RL at a 30% W/V solution was added (900 mg) and blending was continued once there was a homogeneous blend then 50 mL of ethanol was added to the mixture for the purpose of hardening the spheres. The fluid was then placed on a preheated stirring plate and at very low temperature <40°C and stirred 1700 Rpm's for 30 minutes. After stirring was complete, the fluid was placed into a modified Pyrex® dish and allowed to air dry for 24 hours. At the end of this time there was a very fine granulated powder that was removed by a sharp razor blade edge and weighed. The theoretical yield was 2.80 grams, while the actual yield was 2.75 grams.
The spheres were again vortexed in deionized water and protamine was added. There was no evidence of precipitation. However, when acetone and dichloromethane were added a heavy distinct precipitate occurred indicting that the insulin had been entrapped into the nanoparticles. Animal Model
Three (3) test animals, rats made diabetic by streptozocin injection, were given varying amounts of the spheres with a bioadhesive. As this was a trial evaluation, only three animals were tested at this time. All animals had pretest blood sugars of +400 mg/ dL. The results are demonstrated below. This experiment was performed by the veterinary unit at the Colorado State University School Of Veterinary Medicine.
Table 3 Blood Glucose Rat#l O Hour 24 Hours 48 Hours 72 Hours 96 Hours
1092 units 414 ng/dL 212 mg/dL 44 mg/dL 125 mg/dL 386 mg/dL
Rat#2
1092 units 444 mg/dL 420 mg/dL 126 mg/dL 157 mg./dL 328 mg/dL
Rat #3 1092 units 427 mg/dL 312 mg/dL 110 mg/dL 134 mg/dL 470 mg/dL
Although the control and consistency were not as desirable as had been hoped for, the first trial was considered successful in that it was demonstrated yet again that a large molecule can be adrriinistered orally achieving physiologic activity over a prolonged period of time. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity and understanding, it will be obvious to one skilled in the art that certain changes and modifications may be practiced within the scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A controlled release pharmaceutical formulation comprising a biologically active molecule which is formed into nanoparticles with biodegradable hydrogel polymers and then coated with one or more bioadhesive adjuvants.
2. A controlled release pharmaceutical formulation comprising a biologically active molecule which is formed into nanoparticles with biodegradable hydrogel polymers, coated with liposomes, and then coated with one or more bioadhesive adjuvants.
3. A controlled release pharmaceutical formulation comprising a biologically active molecule encapsulated with a cyclodextrin, which is then formed into nanoparticles with biodegradable water-soluble hydrogel polymers, and then coated with one or more bioadhesive adjuvants.
4. A controlled release pharmaceutical formulation comprising a biologically active molecule which is encapsulated with a cyclodextrin, entrapped with liposomes and formed into nanoparticles then with water-soluble hydrogel polymers, and then coated with one or more bioadhesive adjuvants.
5. A controlled release pharmaceutical formulation comprising a biologically active molecule encapsulated with a cyclodextrin which is then formed into nanoparticles with biodegradable water-soluble hydrogel polymers, coated with liposomes, and then coated with one or more bioadhesive adjuvants.
6. The controlled release pharmaceutical formulation of claims 1-5, wherein said biodegradable hydrogel polymer is selected from the group consisting of poly (lactic acid), poly (glycolic acid), (poly (e-caprolactone), poly (p-hydrohybutyrate), poly (β-hydroxyvalerate). poly (tartonic acid), polydixamone, poly (ethylene terephalate) poly (ortho esters), polyanhydrides, dextran, cross-linked polyvinyl alcohol, polyhydroxy methacrylate, polycyanoacrylates, poly (phosphoesters), polyphosphgenes, poly
(hydroxy butyrate co-valerate), poly (2-hydroxyethyl glutamate), polyvinylpyrrolidone, poly (ethylene glycol), poly (propylene glycol), chitosan, pullulan, zein, alginic acid and alginate salts.
7. The controlled release pharmaceutical formulation of claims 1-5, wherein said biodegradable hydrogel polymer is selected from the group consisting of These include, but are not limited to acrylic acid, methacrylacetic cyanoethyl methocrylitic aminoakyl methacrylate copolymers, ethoxyethyl methacrylate copolymers, polymethacrylic acid, methacrylate acid alkylamide copolymer, polymethacrylic acid, polyacrylic acid, methacryic acid alkylamide copolymer, polymethacrylic acid (anhydride), and polymethyl methacrylate.
8. The controlled release pharmaceutical formulation of claims 3-5 wherein said cyclodextrin is selected from alpha, beta, and gamma cyclodextrins or a combination thereof.
9. The controlled release pharmaceutical formulation of claims 2, 4, or 5, wherein said liposomes are selected from the group consisting of cholesterol, lecithin, glycerol monostearate, phospholipon 90H (Natterman) maleated soybean oil, sphingomylein, phosphatidyl entanolamine, dipamitoyl phoisphophatidykaline, phophosinositol, phophatidic acid, triglycerides.
10. The controlled release pharmaceutical formulation of claims 1-5, wherein said bioadhesive adjuvant is a gum.
11. The controlled release pharmaceutical formulation of claim 10, wherein said gum is selected from the group consisting of pectins, guar gum, xantham gum, dextrin, gum acacia, gum dragon, and gum tragacanth.
12. The controlled release pharmaceutical formulation of claims 1-5, wherein said bioadhesive adjuvant is a cellulosic derivative.
13. The controlled release pharmaceutical formulation of claim 12, wherein said cellulosic derivatives is selected from the group consisting of hydroxypropyl cellulose, hydroxymethyl cellulose, hydroxypropyl methyl cellulose, hydroxypropyl alginate, and carboxymethyl cellulose.
14. The controlled release pharmaceutical formulation of claims 1-5, wherein said biologically active molecule is selected from the group consisting of a biological active protein, polypeptide, and/or polysaccharide molecule.
15. The controlled release pharmaceutical formulation of claim 14, wherein said formulation contains more than one biologically active molecule.
16. The controlled release pharmaceutical formulation of claim 14, wherein said biologically active molecule is heparin.
17. The controlled release pharmaceutical formulation of claim 14, wherein said biologically active molecule is insulin.
18. The controlled release pharmaceutical formulation of claims 1-5, wherein said formulation is administered by oral, parenteral or inhalation delivery.
PCT/US1996/012203 1995-07-27 1996-07-25 Drug delivery systems for macromolecular drugs WO1997004747A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US50824795A 1995-07-27 1995-07-27
US08/508,247 1995-07-27

Publications (1)

Publication Number Publication Date
WO1997004747A1 true WO1997004747A1 (en) 1997-02-13

Family

ID=24021960

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/012203 WO1997004747A1 (en) 1995-07-27 1996-07-25 Drug delivery systems for macromolecular drugs

Country Status (1)

Country Link
WO (1) WO1997004747A1 (en)

Cited By (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6524595B1 (en) * 2000-05-12 2003-02-25 Coletica Cyclodextrins preferentially substituted on their primary face by acid or amine functions
WO2003053470A2 (en) * 2001-12-21 2003-07-03 Universiteit Gent Pulsed bio-agent delivery systems based on degradable polymer solutions or hydrogels
EP1385530A1 (en) * 2001-05-09 2004-02-04 Mediplex Corporation Formulation of amphiphilic heparin derivatives for enhancing mucosal absorption
US6740643B2 (en) * 1999-01-21 2004-05-25 Mirus Corporation Compositions and methods for drug delivery using amphiphile binding molecules
US6875448B1 (en) * 1999-09-03 2005-04-05 Chugai Seiyaku Kabushiki Kaisha Method of intracellular sustained-release of drug and preparations
WO2005110374A1 (en) * 2004-04-30 2005-11-24 Allergan, Inc. Intraocular drug delivery systems containing a therapeutic component, a cyclodextrin, and a polymeric component
EP1696961A1 (en) * 2003-12-24 2006-09-06 Samyang Corporation Nanoparticle compositions of water-soluble drugs for oral administration and preparation methods thereof
WO2006128937A2 (en) 2005-06-02 2006-12-07 Universidade De Santiago De Compostela Nanoparticles comprising chitosan and cyclodextrin
KR100690318B1 (en) 2004-09-25 2007-03-09 경북대학교 산학협력단 Process for preparing nano-hydrogel of hydrophilic polymer
EP1774971A1 (en) * 2005-10-14 2007-04-18 Advanced in Vitro Cell Technologies, S.L. Chitosan and heparin nanoparticles
EP1808438A3 (en) * 1999-06-29 2008-12-24 MannKind Corporation Purification and stabilization of peptide and proteins in pharmaceutical agents
WO2011015634A2 (en) 2009-08-05 2011-02-10 Pieris Ag Controlled release formulations of lipocalin muteins
EP2316490A2 (en) 2000-10-31 2011-05-04 PR Pharmaceuticals, Inc. Methods and compositions for enhanced delivery of bioactive molecules
WO2012050359A2 (en) * 2010-10-14 2012-04-19 (주)아모레퍼시픽 Hydrogel particle coated with lipid and method for manufacturing same
WO2013134349A1 (en) 2012-03-06 2013-09-12 Clemson University Delivery systems for enhancing drug efficacy
US8628801B2 (en) 2004-04-29 2014-01-14 Universidad De Navarra Pegylated nanoparticles
US8629151B2 (en) 2009-05-27 2014-01-14 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US8871269B2 (en) 2003-07-15 2014-10-28 Evonik Corporation Method for the preparation of controlled release formulations
US8895067B2 (en) 2004-04-29 2014-11-25 Universidad De Navarra Immune response stimulating composition comprising nanoparticles based on a methyl vinyl ether-maleic acid copolymer
US8900636B2 (en) 2003-07-23 2014-12-02 Evonik Corporation Controlled release compositions
US8906381B2 (en) 2008-10-12 2014-12-09 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IGG humoral response without T-cell antigen
US8916196B2 (en) 2003-04-10 2014-12-23 Evonik Corporation Method for the production of emulsion-based microparticles
CN104257633A (en) * 2014-08-27 2015-01-07 珠海健帆生物科技股份有限公司 Anticoagulant sustain-released capsule, preparation method thereof, and functional anticoagulant sustain-releasing device
US8932595B2 (en) 2008-10-12 2015-01-13 Massachusetts Institute Of Technology Nicotine immunonanotherapeutics
US9006175B2 (en) 1999-06-29 2015-04-14 Mannkind Corporation Potentiation of glucose elimination
US9066978B2 (en) 2010-05-26 2015-06-30 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9080014B2 (en) 2006-05-15 2015-07-14 Massachusetts Institute Of Technology Polymers for functional particles
KR101550941B1 (en) 2010-10-14 2015-09-07 (주)아모레퍼시픽 Lipid coated hydrogel and the process for preparing thereof
US9192675B2 (en) 2008-06-13 2015-11-24 Mankind Corporation Dry powder inhaler and system for drug delivery
US9217129B2 (en) 2007-02-09 2015-12-22 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
US9220687B2 (en) 2008-12-29 2015-12-29 Mannkind Corporation Substituted diketopiperazine analogs for use as drug delivery agents
US9233072B2 (en) 2008-10-12 2016-01-12 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US9233159B2 (en) 2011-10-24 2016-01-12 Mannkind Corporation Methods and compositions for treating pain
US9241903B2 (en) 2006-02-22 2016-01-26 Mannkind Corporation Method for improving the pharmaceutic properties of microparticles comprising diketopiperazine and an active agent
US9267937B2 (en) 2005-12-15 2016-02-23 Massachusetts Institute Of Technology System for screening particles
US9283193B2 (en) 2005-09-14 2016-03-15 Mannkind Corporation Method of drug formulation based on increasing the affinity of crystalline microparticle surfaces for active agents
US9308280B2 (en) 2008-10-12 2016-04-12 Massachusetts Institute Of Technology Targeting of antigen presenting cells with immunonanotherapeutics
US9333179B2 (en) 2007-04-04 2016-05-10 Massachusetts Institute Of Technology Amphiphilic compound assisted nanoparticles for targeted delivery
US9346766B2 (en) 2004-08-20 2016-05-24 Mannkind Corporation Catalysis of diketopiperazine synthesis
US9364619B2 (en) 2008-06-20 2016-06-14 Mannkind Corporation Interactive apparatus and method for real-time profiling of inhalation efforts
US9364436B2 (en) 2011-06-17 2016-06-14 Mannkind Corporation High capacity diketopiperazine microparticles and methods
US9381477B2 (en) 2006-06-23 2016-07-05 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US9474717B2 (en) 2007-10-12 2016-10-25 Massachusetts Institute Of Technology Vaccine nanotechnology
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
US9630930B2 (en) 2009-06-12 2017-04-25 Mannkind Corporation Diketopiperazine microparticles with defined specific surface areas
US9662461B2 (en) 2008-06-13 2017-05-30 Mannkind Corporation Dry powder drug delivery system and methods
US9675674B2 (en) 2004-08-23 2017-06-13 Mannkind Corporation Diketopiperazine salts for drug delivery and related methods
US9700690B2 (en) 2002-03-20 2017-07-11 Mannkind Corporation Inhalation apparatus
US9706944B2 (en) 2009-11-03 2017-07-18 Mannkind Corporation Apparatus and method for simulating inhalation efforts
US9802012B2 (en) 2012-07-12 2017-10-31 Mannkind Corporation Dry powder drug delivery system and methods
US9925144B2 (en) 2013-07-18 2018-03-27 Mannkind Corporation Heat-stable dry powder pharmaceutical compositions and methods
US9943571B2 (en) 2008-08-11 2018-04-17 Mannkind Corporation Use of ultrarapid acting insulin
US9983108B2 (en) 2009-03-11 2018-05-29 Mannkind Corporation Apparatus, system and method for measuring resistance of an inhaler
US10159644B2 (en) 2012-10-26 2018-12-25 Mannkind Corporation Inhalable vaccine compositions and methods
US10307464B2 (en) 2014-03-28 2019-06-04 Mannkind Corporation Use of ultrarapid acting insulin
US10342938B2 (en) 2008-06-13 2019-07-09 Mannkind Corporation Dry powder drug delivery system
US10421729B2 (en) 2013-03-15 2019-09-24 Mannkind Corporation Microcrystalline diketopiperazine compositions and methods
US10561806B2 (en) 2014-10-02 2020-02-18 Mannkind Corporation Mouthpiece cover for an inhaler
US10625034B2 (en) 2011-04-01 2020-04-21 Mannkind Corporation Blister package for pharmaceutical cartridges
US10933129B2 (en) 2011-07-29 2021-03-02 Selecta Biosciences, Inc. Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses
CN114432243A (en) * 2022-01-20 2022-05-06 华南师范大学 Nano-carrier responding to pH and glucose and application thereof
US11446127B2 (en) 2013-08-05 2022-09-20 Mannkind Corporation Insufflation apparatus and methods
US20230165795A1 (en) * 2018-03-20 2023-06-01 Universiteit Twente Liposomes for targeting tumor-associated macrophages

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5346702A (en) * 1992-12-04 1994-09-13 Sterling Winthrop Inc. Use of non-ionic cloud point modifiers to minimize nanoparticle aggregation during sterilization
US5417982A (en) * 1994-02-17 1995-05-23 Modi; Pankaj Controlled release of drugs or hormones in biodegradable polymer microspheres
US5510118A (en) * 1995-02-14 1996-04-23 Nanosystems Llc Process for preparing therapeutic compositions containing nanoparticles

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5346702A (en) * 1992-12-04 1994-09-13 Sterling Winthrop Inc. Use of non-ionic cloud point modifiers to minimize nanoparticle aggregation during sterilization
US5417982A (en) * 1994-02-17 1995-05-23 Modi; Pankaj Controlled release of drugs or hormones in biodegradable polymer microspheres
US5510118A (en) * 1995-02-14 1996-04-23 Nanosystems Llc Process for preparing therapeutic compositions containing nanoparticles

Cited By (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6740643B2 (en) * 1999-01-21 2004-05-25 Mirus Corporation Compositions and methods for drug delivery using amphiphile binding molecules
US9801925B2 (en) 1999-06-29 2017-10-31 Mannkind Corporation Potentiation of glucose elimination
EP1808438A3 (en) * 1999-06-29 2008-12-24 MannKind Corporation Purification and stabilization of peptide and proteins in pharmaceutical agents
US8389470B2 (en) 1999-06-29 2013-03-05 Mannkind Corporation Methods and compositions for delivering peptides
US9006175B2 (en) 1999-06-29 2015-04-14 Mannkind Corporation Potentiation of glucose elimination
US7943178B2 (en) 1999-06-29 2011-05-17 Mannkind Corporation Methods and compositions for delivering peptides
US7648960B2 (en) 1999-06-29 2010-01-19 Mannkind Corporation Method for delivery of monomeric or dimeric insulin complexed to diketopiperazine microparticles
US6875448B1 (en) * 1999-09-03 2005-04-05 Chugai Seiyaku Kabushiki Kaisha Method of intracellular sustained-release of drug and preparations
US6524595B1 (en) * 2000-05-12 2003-02-25 Coletica Cyclodextrins preferentially substituted on their primary face by acid or amine functions
EP2316490A2 (en) 2000-10-31 2011-05-04 PR Pharmaceuticals, Inc. Methods and compositions for enhanced delivery of bioactive molecules
EP1385530A4 (en) * 2001-05-09 2005-11-09 Mediplex Corp Formulation of amphiphilic heparin derivatives for enhancing mucosal absorption
EP1385530A1 (en) * 2001-05-09 2004-02-04 Mediplex Corporation Formulation of amphiphilic heparin derivatives for enhancing mucosal absorption
WO2003053470A3 (en) * 2001-12-21 2004-03-18 Univ Gent Pulsed bio-agent delivery systems based on degradable polymer solutions or hydrogels
WO2003053470A2 (en) * 2001-12-21 2003-07-03 Universiteit Gent Pulsed bio-agent delivery systems based on degradable polymer solutions or hydrogels
US9700690B2 (en) 2002-03-20 2017-07-11 Mannkind Corporation Inhalation apparatus
US10272044B2 (en) 2003-04-10 2019-04-30 Evonik Corporation Method for the production of emulsion-based microparticles
US8916196B2 (en) 2003-04-10 2014-12-23 Evonik Corporation Method for the production of emulsion-based microparticles
US8871269B2 (en) 2003-07-15 2014-10-28 Evonik Corporation Method for the preparation of controlled release formulations
US8900636B2 (en) 2003-07-23 2014-12-02 Evonik Corporation Controlled release compositions
EP1696961A4 (en) * 2003-12-24 2010-09-01 Samyang Corp Nanoparticle compositions of water-soluble drugs for oral administration and preparation methods thereof
EP1696961A1 (en) * 2003-12-24 2006-09-06 Samyang Corporation Nanoparticle compositions of water-soluble drugs for oral administration and preparation methods thereof
US8895067B2 (en) 2004-04-29 2014-11-25 Universidad De Navarra Immune response stimulating composition comprising nanoparticles based on a methyl vinyl ether-maleic acid copolymer
US8628801B2 (en) 2004-04-29 2014-01-14 Universidad De Navarra Pegylated nanoparticles
WO2005110374A1 (en) * 2004-04-30 2005-11-24 Allergan, Inc. Intraocular drug delivery systems containing a therapeutic component, a cyclodextrin, and a polymeric component
US9346766B2 (en) 2004-08-20 2016-05-24 Mannkind Corporation Catalysis of diketopiperazine synthesis
US9796688B2 (en) 2004-08-20 2017-10-24 Mannkind Corporation Catalysis of diketopiperazine synthesis
US9675674B2 (en) 2004-08-23 2017-06-13 Mannkind Corporation Diketopiperazine salts for drug delivery and related methods
US10130685B2 (en) 2004-08-23 2018-11-20 Mannkind Corporation Diketopiperazine salts for drug delivery and related methods
KR100690318B1 (en) 2004-09-25 2007-03-09 경북대학교 산학협력단 Process for preparing nano-hydrogel of hydrophilic polymer
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
WO2006128937A2 (en) 2005-06-02 2006-12-07 Universidade De Santiago De Compostela Nanoparticles comprising chitosan and cyclodextrin
US10143655B2 (en) 2005-09-14 2018-12-04 Mannkind Corporation Method of drug formulation
US9446001B2 (en) 2005-09-14 2016-09-20 Mannkind Corporation Increasing drug affinity for crystalline microparticle surfaces
US9717689B2 (en) 2005-09-14 2017-08-01 Mannkind Corporation Method of drug formulation based on increasing the affinity of crystalline microparticle surfaces for active agents
US9283193B2 (en) 2005-09-14 2016-03-15 Mannkind Corporation Method of drug formulation based on increasing the affinity of crystalline microparticle surfaces for active agents
WO2007042572A1 (en) * 2005-10-14 2007-04-19 Advanced In Vitro Cell Technologies, S.L. Chitosan and heparin nanoparticles
EP1774971A1 (en) * 2005-10-14 2007-04-18 Advanced in Vitro Cell Technologies, S.L. Chitosan and heparin nanoparticles
AU2006301162B2 (en) * 2005-10-14 2012-04-05 Advancell Advanced In Vitro Cell Technologies, S.A. Chitosan and heparin nanoparticles
US9267937B2 (en) 2005-12-15 2016-02-23 Massachusetts Institute Of Technology System for screening particles
US9241903B2 (en) 2006-02-22 2016-01-26 Mannkind Corporation Method for improving the pharmaceutic properties of microparticles comprising diketopiperazine and an active agent
US10130581B2 (en) 2006-02-22 2018-11-20 Mannkind Corporation Method for improving the pharmaceutic properties of microparticles comprising diketopiperazine and an active agent
US9688812B2 (en) 2006-05-15 2017-06-27 Massachusetts Institute Of Technology Polymers for functional particles
US9080014B2 (en) 2006-05-15 2015-07-14 Massachusetts Institute Of Technology Polymers for functional particles
US9381477B2 (en) 2006-06-23 2016-07-05 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US9217129B2 (en) 2007-02-09 2015-12-22 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
US9333179B2 (en) 2007-04-04 2016-05-10 Massachusetts Institute Of Technology Amphiphilic compound assisted nanoparticles for targeted delivery
US9474717B2 (en) 2007-10-12 2016-10-25 Massachusetts Institute Of Technology Vaccine nanotechnology
US9526702B2 (en) 2007-10-12 2016-12-27 Massachusetts Institute Of Technology Vaccine nanotechnology
US11547667B2 (en) 2007-10-12 2023-01-10 Massachusetts Institute Of Technology Vaccine nanotechnology
US9539210B2 (en) 2007-10-12 2017-01-10 Massachusetts Institute Of Technology Vaccine nanotechnology
US10736848B2 (en) 2007-10-12 2020-08-11 Massachusetts Institute Of Technology Vaccine nanotechnology
US9339615B2 (en) 2008-06-13 2016-05-17 Mannkind Corporation Dry powder inhaler and system for drug delivery
US10201672B2 (en) 2008-06-13 2019-02-12 Mannkind Corporation Dry powder inhaler and system for drug delivery
US10751488B2 (en) 2008-06-13 2020-08-25 Mannkind Corporation Dry powder inhaler and system for drug delivery
US10342938B2 (en) 2008-06-13 2019-07-09 Mannkind Corporation Dry powder drug delivery system
US9662461B2 (en) 2008-06-13 2017-05-30 Mannkind Corporation Dry powder drug delivery system and methods
US9511198B2 (en) 2008-06-13 2016-12-06 Mannkind Corporation Dry powder inhaler and system for drug delivery
US9446133B2 (en) 2008-06-13 2016-09-20 Mannkind Corporation Dry powder inhaler and system for drug delivery
US9192675B2 (en) 2008-06-13 2015-11-24 Mankind Corporation Dry powder inhaler and system for drug delivery
US9364619B2 (en) 2008-06-20 2016-06-14 Mannkind Corporation Interactive apparatus and method for real-time profiling of inhalation efforts
US10675421B2 (en) 2008-06-20 2020-06-09 Mannkind Corporation Interactive apparatus and method for real-time profiling of inhalation efforts
US9943571B2 (en) 2008-08-11 2018-04-17 Mannkind Corporation Use of ultrarapid acting insulin
US9233072B2 (en) 2008-10-12 2016-01-12 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8932595B2 (en) 2008-10-12 2015-01-13 Massachusetts Institute Of Technology Nicotine immunonanotherapeutics
US9439859B2 (en) 2008-10-12 2016-09-13 Massachusetts Institute Of Technology Adjuvant incorporation in immunoanotherapeutics
US8906381B2 (en) 2008-10-12 2014-12-09 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IGG humoral response without T-cell antigen
US9308280B2 (en) 2008-10-12 2016-04-12 Massachusetts Institute Of Technology Targeting of antigen presenting cells with immunonanotherapeutics
US9220687B2 (en) 2008-12-29 2015-12-29 Mannkind Corporation Substituted diketopiperazine analogs for use as drug delivery agents
US10172850B2 (en) 2008-12-29 2019-01-08 Mannkind Corporation Substituted diketopiperazine analogs for use as drug delivery agents
US9655850B2 (en) 2008-12-29 2017-05-23 Mannkind Corporation Substituted diketopiperazine analogs for use as drug delivery agents
US9983108B2 (en) 2009-03-11 2018-05-29 Mannkind Corporation Apparatus, system and method for measuring resistance of an inhaler
US8629151B2 (en) 2009-05-27 2014-01-14 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US9884112B2 (en) 2009-05-27 2018-02-06 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US9006254B2 (en) 2009-05-27 2015-04-14 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US9630930B2 (en) 2009-06-12 2017-04-25 Mannkind Corporation Diketopiperazine microparticles with defined specific surface areas
EP2550958A1 (en) 2009-08-05 2013-01-30 Pieris AG Controlled release formulations of lipocalin muteins
WO2011015634A2 (en) 2009-08-05 2011-02-10 Pieris Ag Controlled release formulations of lipocalin muteins
US9706944B2 (en) 2009-11-03 2017-07-18 Mannkind Corporation Apparatus and method for simulating inhalation efforts
US9764031B2 (en) 2010-05-26 2017-09-19 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9066978B2 (en) 2010-05-26 2015-06-30 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9849089B2 (en) 2010-10-14 2017-12-26 Amorepacific Corporation Hydrogel particle coated with lipid and method for manufacturing same
WO2012050359A2 (en) * 2010-10-14 2012-04-19 (주)아모레퍼시픽 Hydrogel particle coated with lipid and method for manufacturing same
WO2012050359A3 (en) * 2010-10-14 2012-07-05 (주)아모레퍼시픽 Hydrogel particle coated with lipid and method for manufacturing same
KR101550941B1 (en) 2010-10-14 2015-09-07 (주)아모레퍼시픽 Lipid coated hydrogel and the process for preparing thereof
CN103167868A (en) * 2010-10-14 2013-06-19 株式会社爱茉莉太平洋 Hydrogel particle coated with lipid and method for manufacturing same
US10625034B2 (en) 2011-04-01 2020-04-21 Mannkind Corporation Blister package for pharmaceutical cartridges
US9364436B2 (en) 2011-06-17 2016-06-14 Mannkind Corporation High capacity diketopiperazine microparticles and methods
US10130709B2 (en) 2011-06-17 2018-11-20 Mannkind Corporation High capacity diketopiperazine microparticles and methods
US10933129B2 (en) 2011-07-29 2021-03-02 Selecta Biosciences, Inc. Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses
US10258664B2 (en) 2011-10-24 2019-04-16 Mannkind Corporation Methods and compositions for treating pain
US9610351B2 (en) 2011-10-24 2017-04-04 Mannkind Corporation Methods and compositions for treating pain
US9233159B2 (en) 2011-10-24 2016-01-12 Mannkind Corporation Methods and compositions for treating pain
WO2013134349A1 (en) 2012-03-06 2013-09-12 Clemson University Delivery systems for enhancing drug efficacy
US9802012B2 (en) 2012-07-12 2017-10-31 Mannkind Corporation Dry powder drug delivery system and methods
US10159644B2 (en) 2012-10-26 2018-12-25 Mannkind Corporation Inhalable vaccine compositions and methods
US10421729B2 (en) 2013-03-15 2019-09-24 Mannkind Corporation Microcrystalline diketopiperazine compositions and methods
US9925144B2 (en) 2013-07-18 2018-03-27 Mannkind Corporation Heat-stable dry powder pharmaceutical compositions and methods
US11446127B2 (en) 2013-08-05 2022-09-20 Mannkind Corporation Insufflation apparatus and methods
US10307464B2 (en) 2014-03-28 2019-06-04 Mannkind Corporation Use of ultrarapid acting insulin
CN104257633A (en) * 2014-08-27 2015-01-07 珠海健帆生物科技股份有限公司 Anticoagulant sustain-released capsule, preparation method thereof, and functional anticoagulant sustain-releasing device
US10561806B2 (en) 2014-10-02 2020-02-18 Mannkind Corporation Mouthpiece cover for an inhaler
US20230165795A1 (en) * 2018-03-20 2023-06-01 Universiteit Twente Liposomes for targeting tumor-associated macrophages
CN114432243A (en) * 2022-01-20 2022-05-06 华南师范大学 Nano-carrier responding to pH and glucose and application thereof
CN114432243B (en) * 2022-01-20 2023-08-29 华南师范大学 Nanocarrier responding to pH and glucose and application thereof

Similar Documents

Publication Publication Date Title
WO1997004747A1 (en) Drug delivery systems for macromolecular drugs
JP5175017B2 (en) Particulate carrier for improving oral absorption of active ingredients
EP1255534B1 (en) Method for the preparation of microspheres which contain colloidal systems
Gan et al. Chitosan nanoparticle as protein delivery carrier—systematic examination of fabrication conditions for efficient loading and release
CA2113243C (en) Pharmaceutical compositions containing nanocapsules
Panos et al. New drug delivery systems based on chitosan
JPH11130697A (en) Pharmaceutical composition controlled in rate for releasing medicine
JPS6133112A (en) Biologically degradable microcapsule and manufacture
JP2009256383A (en) Encapsulation of nanosuspension in liposome and microsphere
Zhang et al. pH-sensitive O-carboxymethyl chitosan/sodium alginate nanohydrogel for enhanced oral delivery of insulin
Pavanetto et al. Evaluation of process parameters involved in chitosan microsphere preparation by the o/w/o multiple emulsion method
EP1371364A1 (en) Proces for the preparation of a controlled release system
US20100143484A1 (en) Oral submicron particle delivery system for proteins and process for its production
Prado et al. Characterization of pcl and chitosan nanoparticles as carriers of enoxaparin and its antithrombotic effect in animal models of venous thrombosis
Jiao et al. Preparation and in vitro evaluation of heparin-loaded polymeric nanoparticles
Sabra et al. Gastrointestinal delivery of APIs from chitosan nanoparticles
WO2002041829A2 (en) Oral nanosphere delivery
CN114522150B (en) Preparation method and application of pH-sensitive plant microcapsule nano extruder
CN109662955A (en) A kind of chitosan drug-loading nano particle of oleanolic acid grafting and its preparation and application
CN1198100A (en) Chitosan drug delivery system
Shivani et al. Colon delivery of 5-fluoro uracil using cross-linked chitosan microspheres coated with eudragit S 100
CN1410056A (en) Preparation method of water soluble anticancer medical microsphere
CN102406610B (en) Particle dosing system with long circulation performance and preparation method thereof
Yang et al. Multifunctional β-Cyclodextrin–Poly (ethylene glycol)–Cholesterol Nanomicelle for Anticancer Drug Delivery
CN115282127B (en) Chitosan calcium alginate nanoparticle microsphere and preparation method thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP MX

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97507726

Format of ref document f/p: F

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA