WO1993025583A2 - Proteinoid carriers and methods for preparation and use thereof - Google Patents

Proteinoid carriers and methods for preparation and use thereof Download PDF

Info

Publication number
WO1993025583A2
WO1993025583A2 PCT/US1993/005723 US9305723W WO9325583A2 WO 1993025583 A2 WO1993025583 A2 WO 1993025583A2 US 9305723 W US9305723 W US 9305723W WO 9325583 A2 WO9325583 A2 WO 9325583A2
Authority
WO
WIPO (PCT)
Prior art keywords
proteinoid
group
acid
carrier
carriers
Prior art date
Application number
PCT/US1993/005723
Other languages
French (fr)
Other versions
WO1993025583A3 (en
WO1993025583A9 (en
Inventor
Sam J. Milstein
Martin L. Kantor
Original Assignee
Emisphere Technologies, Inc.
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
Priority claimed from US07/920,346 external-priority patent/US5443841A/en
Application filed by Emisphere Technologies, Inc. filed Critical Emisphere Technologies, Inc.
Priority to KR1019940704564A priority Critical patent/KR950701939A/en
Priority to BR9306678A priority patent/BR9306678A/en
Priority to JP6501793A priority patent/JPH07508004A/en
Priority to AU46356/93A priority patent/AU4635693A/en
Priority to EP93916542A priority patent/EP0642532A1/en
Publication of WO1993025583A2 publication Critical patent/WO1993025583A2/en
Publication of WO1993025583A9 publication Critical patent/WO1993025583A9/en
Publication of WO1993025583A3 publication Critical patent/WO1993025583A3/en
Priority to NO944852A priority patent/NO944852L/en
Priority to FI945912A priority patent/FI945912A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/64Proteins; Peptides; Derivatives or degradation products thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/02Cosmetics or similar toiletry preparations characterised by special physical form
    • A61K8/0241Containing particulates characterized by their shape and/or structure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/167Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction with an outer layer or coating comprising drug; with chemically bound drugs or non-active substances on their surface
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q13/00Formulations or additives for perfume preparations
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/40Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino groups bound to carbon atoms of at least one six-membered aromatic ring and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/42Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino groups bound to carbon atoms of at least one six-membered aromatic ring and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton with carboxyl groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by saturated carbon chains
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/45Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • C07C233/53Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring
    • C07C233/55Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring having the carbon atom of the carboxamide group bound to a carbon atom of an unsaturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/57Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C233/63Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/64Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C233/81Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • C07C233/82Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/87Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom of a carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/32Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • C07C235/38Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/42Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/44Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/58Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring with carbon atoms of carboxamide groups and singly-bound oxygen atoms, bound in ortho-position to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/64Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring with carbon atoms of carboxamide groups and singly-bound oxygen atoms, bound in ortho-position to carbon atoms of the same non-condensed six-membered aromatic ring having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/70Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/84Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton with the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/22Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/18Systems containing only non-condensed rings with a ring being at least seven-membered
    • 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
    • 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
    • Y10S977/907Liposome
    • 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
    • Y10S977/915Therapeutic or pharmaceutical composition
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]
    • Y10T428/2984Microcapsule with fluid core [includes liposome]
    • Y10T428/2985Solid-walled microcapsule from synthetic polymer
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]
    • Y10T428/2989Microcapsule with solid core [includes liposome]

Definitions

  • This invention relates to proteinoids and proteinoid carriers made from them.
  • the proteinoid carriers releasably encapsulate active agents and have extended longer shelf life and/or photostability. Methods for the preparation of such proteinoid carriers are also disclosed.
  • adjuvants such as resorcinols and non- ionic surfactants polyoxyethylene oleyl ether and n-hexadecyl polyethylene ether to increase the permeability of the intestinal walls; and
  • enzymatic inhibitors such as pancreatic trypsin inhibitor, diisopropylfluorophosphate (DFF) and nrasylol to avoid enzymatic degradation.
  • Liposomes as drug delivery systems have also been described. They provide a layer of lipid around the encapsulated pharmacological agent.
  • the use of liposomes containing heparin is disclosed in U.S. Patent No. 4,239,754 and several studies have been directed co the use of liposomes containing insulin; e.g., Patel et al. (1976) FEBS Letters Vol. 62, page 60 and Hashimoto et al. (1979) Endocrinol. Japan, Vol. 26, page 337.
  • the use of liposomes is still in the development stage and there are continuing problems, including:
  • microspheres More recently, synthetic amino acid polymers or proteinoids, forming microspheres, have been described for encapsulating pharmaceuticals.
  • U.S. Patent No. 4,925,673 (the '673 patent), the disclosure which is hereby incorporated by reference in its entirety, describes such microsphere constructs ai-well as methods for their preparation and use.
  • the '673 patent also describe: microspheres which encapsulate pharmaceuti-cal agents for delivery into the gastrointestinal tract or into the blood.
  • proteinoid microspheres described in the '673 patent are useful for their intended purposes, the physicochemical properties of the proteinoid microspheres, such as light sensitivity, shelf life and the selectivity of their solubility in various portions of the gastrointestinal tract, could be improved. Additionally, there is a need in the art for microspheres that can encapsulate a broader range of active agents such as polar drugs.
  • MW (MW) (> 1000 daltons) and low MW ( ⁇ 1000 daltons) peptide-like polymers which are difficult to separate.
  • the method produces a small amount of the low MW proteinoids which is the microsphere-forming fraction.
  • an improved method of preparing of the proteinoids is also desired.
  • the present invention relates to improved proteinoid carriers and methods of making and use thereof.
  • Proteinoids of a MW ranging between about 250 and about 2400 daltons and of defined amino acids are useful in preparing proteinoid carriers with improved stability against photodegradation and/or decomposition.
  • the proteinoids comprise a peptide polymer selected from the group consisting of:
  • peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid;
  • peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and from at least one third monomer selected from the group consisting of lysine, arginine and ornithine, the proteinoid being a microsphere- and/or microcap- sule-forming proteinoid and being soluble within a selected pH range.
  • the proteinoid molecules of the invention contain between about 2 and about 20 amino acid residues, preferably between about 2 and about 8 amino acid residues, and has a molecular weight which ranges between about 250 and about 2400 daltons, preferably between about 250 and about 600, and most preferably between about 250 and 400 daltons.
  • the proteinoid carriers are useful as delivery systems to releasably encapsulate and carry a broad range of cargoes including pharmaceutical agents, dye reagents and cosmetic ingredients.
  • the proteinoid carriers are useful as oral delivery systems of sensitive pharmaceutical agents, which normally would not be admmistrable via the oral ro ⁇ te for selective release at targeted regions of the gastrointestinal tract.
  • Figure 1 illustrates the molecular weight distribution as a function of monomer concentration of poly (Asp.Bz-co-Phe) polymer prepared by the NCA method as described in Example 3.
  • Figure 2 illustrates the molecular weight distribution of a function of monomer concentration of poly (Asp.Bz) polymer prepared by the DPPA method as described in Example 5.
  • Figure 3 illustrates the effect of reaction time duration on yields of poly (Asp.Bz) polymer prepared by the DPPA method as described in Example 5.
  • Figure 4 illustrates be effect of temperature of the molecular weight of poly (Asp.Bz) polymer prepared by the DPPA method as described in Example 5.
  • Figure 5 illustrates the effect of changing the molar ratios of [DPPAl/ [M] on the molecular weight of poly (Asp.Bz) polymer by the DPPA method as described in Example 5.
  • Figure 6 is a photograph of an x-ray film of the western immunoblot analysis, as described in Example 9, of purified murine mAb 9BG5 (2 ⁇ g, lane 1; lmg, lane 2; and 0.25 ⁇ g, lane 3); empty proteinoid carrier supernatant after encapsulating process (no mAb) (lane 4); empty proteinoid carrier pellet (lane 5); proteinoid carrier encapsulated mAb supernatant after encapsulating process (lane 6); and proteinoid carrier encapsulated mAb pellet.
  • Lane MW contained standard molecular weight markers.
  • Figure 7 is a photograph of an x-ray film of a western immunoblot analysis of samples described in Example 10.
  • Figures 8 (a-c) illustrate the levels of serum proteins which bound to immobilized reovirus type 3 and V L SH under ELISA conditions as described in Example 11.
  • "Empty spheres” refers to animals orally administered empty proteinoid carriers (no mAb 9BG5);
  • mAb spheres refers to animals orally administered mAb 9BG5 encapsulated proteinoid carriers;
  • IV refers to animals intravenously administered unencapsulated mAb 9BG5; and
  • oral refers to animals orally administered unencapsulated mAb 9BG5.
  • Figure 9 show mAb binding under conventional ELISA procedures using immobilized reovirus type 3 and V L SH proteins with serial dilutions of purified mAb in 0.85 N citrate-0.5% gum ( Figure 9 (a)) or phosphate buffered saline ( Figure 9 (b)) as described in Example 11.
  • Figure 10 illustrates levels of erythropoietin (EPO) detected in rat serum taken from rats administered proteinoid carrier encapsulated EPO (15 ⁇ g EPO/kg body weight) and encapsulated EPO (15 ⁇ g EPO/kg body weight) as described in Example 15.
  • EPO erythropoietin
  • Figure 11 illustrates EPO serum levels in rats that were administered either erythropoietin (50 ⁇ g/kg) or encapsulated erythropoietin (50 ⁇ g/kg) directly into the proximal duodenum as described in Example 15. Serum erythropoietin levels were determined over time with a erythropoietin enzyme immunoassay kit.
  • Figure 12 illustrates EPO serum levels in rats who were orally gavaged with either encapsulated or unencapsulated erythropoietin (100 ⁇ g/kg) or received a subcutaneous injection of either 2 ⁇ g/kg or 10 ⁇ g/kg as described in Example 15. Serum erythropoietin levels were determined over time with an erythropoietin enzyme immunoassay kit.
  • Figure 13 illustrates serum calcium changes after oral administration of salmon calcitonin proteinoid carriers (0.25 mg calcitonin/kg body weight) in cynomolgus monkeys as described in Example 17. The results are expressed as absolute change in serum calcium from baseline values. The data represents means +/- SEM. ** Serum calcium levels significally different from baseline values.
  • Figure 14 illustrates serum calcium changes following oral administration of salmon calcitonin proteinoid carriers (0.60 mg/kg body weight) in rats as described in Example 18. The results are expressed as absolute change in serum calcium from baseline values. The data represents means +/- SEM. **Serum calcium levels significantly different compared to the control group at the corresponding time point.
  • Figure 15 illustrate, serum calcium changes after intraduodenal administration of salmon calcitonin or calcitonin proteinoid carriers (3 ug/kg body weight) in rats as described in Example 18. The results are expressed as absolute change in serum calcium from baseline values. The data represents means +/- SEM. ** Significantly different from the unencapsulated control group at the indicated time points.
  • Figure 16 illustrates clotting times after oral administration of proteinoid carrier encapsulated Factor IX (FIX sph PO) and IV administration of FIX solution (FIX IV) as described in Example 20.
  • FIX sph PO proteinoid carrier encapsulated Factor IX
  • FIX IV FIX solution
  • Figure 17 illustrates clotting times after oral administration of proteinoid carrier encapsulated Factor IX (FIX sph PO) and FIX solution (FIX unencap PO) or IV administration of FIX solution (FIX IV) as described in Example 21.
  • FIX sph PO proteinoid carrier encapsulated Factor IX
  • FIX unencap PO FIX solution
  • IV FIX solution
  • Figure 18 illustrates the percentage of intact alpha- interferon (IFN) remaining after incubating IFN and IFN proteinoid carriers in simulated gastric fluid (SGF).
  • Figure 19 illustrates the percentage of intact IFN remaining after incubating IFN and IFN proteinoid carriers in 0.08N HCl.
  • IFN alpha- interferon
  • Figure 20 illustrates the percentage of intact IFN remaining after incubating IFN and IFN proteinoid carriers in simulated intestinal fluid (SIF).
  • Figure 21 illustrates the clotting times in rats dosed with heparin or proteinoid/heparin, both in water.
  • the data represents an average of 6 rats.
  • the data represents means +/- SEM.
  • Figure 22 illustrates clotting times in rats dosed ID with USP heparin or heparin proteinoid carriers, both in citric acid. Each time point is an average of 12 rats. The data represents means +/- SEM.
  • Figure 23 illustrates clotting times in rats dosed orally with heparin-spiked empty proteinoid carriers or heparin proteinoid carriers. Each time point is an average of 12 rats. The data represents means +/- SEM.
  • Figure 24 illustrates the average titers of rats immunized orally with M1 proteinoid carriers versus unencapsulated M1. Only responders in each group were averaged.
  • Figure 25 illustrates HA-NA titers of rats immunized orally with HA-NA micropspheres versus unencapsulated HA-NA.
  • the instant invention arose from the discovery that proteinoids of a MW of between about 250 and about 2400 daltons and of defined amino acid composition can be obtained by modifying known reactions and selecting starting materials. These proteinoids form proteinoid carriers with surprisingly enhanced stability against at least one of photodegradation and decomposition over time.
  • proteinoid carriers prepared from such proteinoids carr. a broader range of pharmaceutical agents, including labile polypeptides such as insulin, alpha-interferon, calcitonin, antigens, e.g. influenza virus M1-protein, and Factor IX and display a selective releasability within various portions of the gastrointestinal tract, relative to prior art proteinoid microspheres.
  • the proteinoids of the invention comprise a peptide polymer selected from the group consisting of:
  • peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid;
  • peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and from at least one third monomer selected from the group consisting of lysine, arginine and ornithine, the proteinoid being a microsphere- or microcapsule-forming proteinoid and being soluble within a selected pH range.
  • the proteinoid molecules of the invention contain between about 2 and about 20 amino acid residues, preferably between about 2 and about 8 amino acid residues, and have a molecular weight which ranges between 250 and about 2400 daltons, preferably between about 250 and about 600, and most preferably between about 250 and 400 daltons.
  • Proteinoid carriers prepared from the proteinoid molecules, in accordance with the present invention display a selective solubility at specific acidic or basic pH ranges, depending on the choice and amount of the second and third monomers in the proteinoid.
  • Proteinoid carriers which are selectively soluble under alkaline pH environments sue are prepared from base-soluble proteinoids. These proteinoids contain, as starting monomers in the reaction mixture, at least one second monomer selected from the group consisting of glutamic acid, glutamine, pyroglutamic acid, and aspartic acid. At a pH ranging between about 7.2 and about 11.0, the base-soluble proteinoid exists largely as the anion and is soluble. At a pH below about 7.0, the proteinoid is largely protonated and insoluble in water.
  • proteinoid carriers which are selectively soluble under acidic pH environments, such as the stomach, are prepared from acid-soluble proteinoids.
  • the proteinoid contain, as starting monomers in the proteinoid reaction mixture, at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid and at least one third monomer selected from the group consisting of lysine, arginine, and ornithine.
  • the base-soluble proteinoid exists largely as the cation and is soluble.
  • the proteinoid is largely unprotonated and insoluble in water.
  • the pH and the solubility characteristics of the acid-soluble proteinoid depends largely, but not exclusively, upon the pH and solubilty of the last amino acid added during the synthesis of the proteinoid.
  • a basic amino acid e.g., a third monomer, selected from the group consisting of lysine, arginine and ornithine in the acid-soluble proteinoid will result in the elevation of the pI (pH at the isoelectric point) of the proteinoid.
  • the proteinoids of the present invention are preparable by a thermal condensation reaction by heating mixtures of the appropriate amino acids under conditions described in the '673 patent.
  • mixtures of two to five specific amino acids with at least one selected from each of the aforementioned groups yield proteinoids which form proteinoid carriers with selective solubility at particular pH ranges and at nigh yields.
  • individual amino acids are added to a reaction flask containing tetramethyl the sulfone (sulfolane) wnich has been heated to a temperatur ranging bet en about 130°C and about 200°C, preferably about 175°C to 195°C under an inert atmosphere of argon or nitrogen gas. After each addition, the solution is stirred for a period of time ranging between about 10 minutes id about hours, depending on the amino acid type and the order of addition.
  • the NCA method involves the preparation of N-carboxyanhydrides of alpha-amino acid esters and their subsequent polymerization, using low MW amines as initiators. It has been discovered that non-NCA derived amino esters, e.g., ⁇ -methyl tyrosine ester, are effective initiators which are stable and soluble in many organic solvents such as tetrahydrofuran (THF). The use of amino acids as initiators, presumably due to their poor solubility in organic solvents and their low stability, are not known. The NCA reaction produces a high yield of proteinoids with high purity.
  • THF tetrahydrofuran
  • the DPPA method involves the direct condensation of benzyl esters of alpha-amino acids in the presence of DPPA and a low MW amine, followed by removal of the protective benzyl groups, contained in the proteinoid product, by alkaline hydrolysis. If catalytic hydrogenation is used in place of alkaline hydrolysis, low MW proteinoids of unexpected high purities and yields are obtained.
  • Proteinoids prepared by any of the above methods can be used immediately to microencapsulate an active pharmacological agent or the proteinoid can be concentrated or dried by conventional means and stored for future use.
  • the proteinoids of the invention are purified as follows: crude proteinoids are slurried with water at room temperature, e.g. 25°C. While at this temperature, the pH of the slurry is adjusted to about pH 8 using an aqueous alkaline solution, e.g. 40% sodium hydroxide and 10% sodium bicarbonate solutions for an acid-soluble proteinoid. For a base-soluble proteinoid, the slurry is adjusted to an acidic pH with an aqueous acidic solution, e.g. 10% acetic acid solution. The mixture is then filtered and the filter cake washed with a volume of water. The washes and filtrate are then combined and evaporated to dryness in vacuo to afford proteinoids. If necessary, this process can be repeated until proteinoids of a desired purity level are obtained.
  • an aqueous alkaline solution e.g. 40% sodium hydroxide and 10% sodium bicarbonate solutions for an acid-soluble proteinoid.
  • the slurry is adjusted to an acidic pH with an
  • the proteinoid may be further purified by fractionating on a column containing solid supports which include silica gel or alumina, using methanol or propanol as mobile phase; ion exchange resin using water as the mobile phase; reverse phase column supports using trifluoroacetic acid/acetonitrile mixtures as mobile phase.
  • the proteinoids may also be purified by extraction with a lower alcohol such as propanol or butanol to remove low molecular weight contaminants.
  • Proteinoid carriers are made from purified proteinoids as follows: proteinoids are dissolved in deionized water at a concentration ranging between about 75 and about 200 mg/ml, preferably about 100 mg/ml, at a temperature between about 25oC and about 60oC, preferably about 40°C. Particulates remaining in the solution may be filtered out by conventional means such as gravity filtration over filter paper.
  • the proteinoid solution maintained at a temperature of about 40°C, is mixed with an aqueous acid solution (also at about 40°C) having an acid concentration ranging between about 1 N and about 2 N, preferably about 1.7 N.
  • the resulting mixture is further incubated at 40°C for a period of time effective for microsphere and microcapsule formation as observed by light microscopy.
  • the preferred order of addition is adding the proteinoid solution to the aqueous acid solution.
  • Suitable acids include any acid which does not (a) adversely effect the proteinoid, e.g., chemical decomposition;
  • Preferred acids for use in this invention include acetic acid, citric acid, hydrochloric acid, phosphoric acid, malic acid and maleic acid.
  • a proteinoid carrier stabilizing additives are preferably incorporated into the aqueous acid solution or into the proteinoid solution, prior to the microsphere or microcapsule formation process.
  • the presence of such additives promotes the stability and dispersibility of the proteinoid carriers in solution.
  • the additives may be employed at a concentration ranging between about 0.1 and 5 % (W/V), preferably about 0.5 % (W/V).
  • Suitable, but non-limiting, examples of stabilizing additives include gum acacia, gelatin, polyethylene glycol, and polylysine.
  • the proteinoid carriers may be used immediately or may be stored at 4oC or lyophilized and stored under desiccant at room temperature or below.
  • the proteinoid molecules form spherical proteinoid carriers comprising proteinoid microcapsules and proteinoid microspheres of less than 10 micron diameter.
  • a-"microsphere is spherical homogeneous mesh work structure having no discrete inner chamber.
  • a “microcapsule” refers to a spherical structure having a proteinoid wall which forms a hollow or chamber.
  • the proteinoid carriers are formed in the presence of a soluble material, e.g., a pharmaceutical agent in the aforementioned aqueous acid solution, this material is believed to be encapsulated within the hollows of the microcapsules and confined within the proteinoid wall defined by the spherical structure or entrapped within the matrix of proteinoid molecules in the microsphere structure.
  • a soluble material e.g., a pharmaceutical agent in the aforementioned aqueous acid solution
  • this material is believed to be encapsulated within the hollows of the microcapsules and confined within the proteinoid wall defined by the spherical structure or entrapped within the matrix of proteinoid molecules in the microsphere structure.
  • pharmacologically active materials such as peptides, proteins, and polysaccharides as well as charged organic molecules, e.g., quinolones or antimicrobial agents, having poor bioavailability by the oral route.
  • the proteinoid carriers of the invention are pharmacologically harmless and do not alter the physiological and biological properties of the active agent. Furthermore, the encapsulation pre ess does not alter the pharmacological properties of the active agent. While any suitable pharmacological agent can be encapsulated within proteinoid carriers, it is particularly valuable for delivering agents which otherwise would be destroyed or rendered less effective by conditions encourtered in the animal body before it reaches its target zone and which are poorly absorbed in the gastrointestinal tract.
  • the proteinoid carriers of the invention are particularly useful for the oral administration of certain pharmacological agents, e.g., small peptide hormones, swhich, by themselves, pass slowly or not at all through the gastro-intestinal mucosa and/or are. susceptible to chemical cleava ⁇ e by acids and enzymes in the gastrointestinal tract.
  • pharmacological agents e.g., small peptide hormones, swhich, by themselves, pass slowly or not at all through the gastro-intestinal mucosa and/or are. susceptible to chemical cleava ⁇ e by acids and enzymes in the gastrointestinal tract.
  • agents include human or bovine growth hormone, interferon and interleukin-II, calcitonin, atrial naturetic factor, antigens, monoclonal antibodies, and Factor IX,. a vitamin K-dependent blood coagulation proenzyme.
  • proteinoids made from gl; tamic acid, aspartic acid, tyrosine, and phenylalanine are especially suitable for encapsulating polysaccharides like heparin.
  • the particle size of the proteinoid carrier plays an important role in determining release of the active agent in the targeted area of the gastrointestinal tract.
  • Proteinoid carriers having diameters between about ⁇ 0.1 microns and about 10 microns, preferably between about 5.0 microns and about 0.1 microns, and containing encapsulated or entrapped active agents are sufficiently small to effectively release the active agent at the targeted area within the gastrointestinal tract.
  • Large proteinoid carriers (>10 microns) tend to be less effective as oral delivery systems.
  • the size of the proteinoid carriers formed by contacting proteinoids with water or aqueous solution containing active agents can be controlled by manipulating a variety of physical or chemical parameters, such as the pH, osmolarity or salt content of the encapsulating solution, and the choice of acid used in the encapsulating process.
  • active agent bearing proteinoid carriers can be produced from base-soluble proteinoids which are stable in the highly acidic stomach (normal pH of from about 2 to about 6), but which dissolve in the distal portion of the intestines.
  • base-soluble proteinoids which are stable in the highly acidic stomach (normal pH of from about 2 to about 6), but which dissolve in the distal portion of the intestines.
  • Such systems are suitable for oral administration of peptide hormones, e.g., insulin, and polysaccharides, e.g., heparin, which otherwise would be quickly destroyed in the GI tract. They also are suitable for protecting the stomach from gastric irritants, such as aspirin.
  • aspirin-containing proteinoid carriers When such aspirin-containing proteinoid carriers are orally administered, they pass through the gastrointestinal mucosa and release the aspirin far more rapidly than conventional enterically coated aspirin, which first must traverse the stomach and then must enter the bloodstream from the intestine after the enteric coating has dissolved.
  • the proteinoid carriers of the invention may be orally administered alone as solids in the form of tablets, pellets, capsules, and granulates suitable for suspension in liquids such as edible oils.
  • the proteinoid carriers can be formulated into an orally administrable composition containing one or more physiologically ..ompatible carriers or excipients.
  • These compositions may contain conventional ingredients such as gelatin, polyvinylpyrrolidone and fillers such as starch and methyl cellulose.
  • the proteinoid carriers of the invention may also be administered by injection.
  • Example 1 Preparation of a Base-soluble Proteinoid
  • the pH of the slurry (at 25°C) was adjusted to 8 using 40% sodium hydroxide solution. The mixture was filtered and the cake washed with a small amount of water. The washes and filtrate are combined and evaporated to dryness in vacuo to give Glu/Asp/Tyr/Phe proteinoid.
  • tetramethylene sulfone 750 ml of tetramethylene sulfone is heated to 190°C in an inert nitrogen atmosphere in a 4 liter flask with stirring.
  • 294 g of glutamic acid is added and the mixture is heated for one-half hour.
  • 362 g of tyrosine is added and the mixture is heated at 190°C for 3 hours.
  • 330 g of phenylalanine is added and the mixture is heated at 190°C for 1.5 hours.
  • 266 g of arginine is added and the mixture is heated for an additional 1.5 hours.
  • the hot melt is then poured into 5 liters of water with vigorous stirring. After stirring for about 1 hour, the mixture is filtered and the filtrate is discarded.
  • the cake is reslurried in 5 liters of water, filtered and the cake is again reslurried in 5 liters of water.
  • the pH of the slurry (at 25°C) was adjusted to 5 using 10% acetic acid solution.
  • the mixture is filtered and the cake is washed with a small amount of water.
  • Appendices A, B, and C describe examples of other proteinoids prepared by the thermocondensation method.
  • This example illustrates the NCA method for preparing copolypeptides consisting of Asp.Bz, Glu.Bz, Phe, and Tyr components.
  • the NCA monomers of these amino acids were prepared according to the reported method.
  • Polydispersity is defined herein as the molecular weight distribution of a sample. The distribution is assigned a numerical value derived from the molecular weight (MW) divided by the molecular number (Mn).
  • the polydispersity value for a homopolymer is 1 because the molecular weight is equal to the molecular number. Any polymer with a polydispersity value of 1 is considered to have a very narrow distribution. A polymer with polydispersity value of 1.6 to 1.7 is considered to have medium distribution. A polymer with a polydispersity value of 2.0-2.1 is considered to have a broad distribution.
  • This example illustrates the method of conducting NCA polymerizations, using ce-methyl tyrosine ester (Tyr.Me) as the initiator.
  • the reaction conditions are essentially the same as described in Example 4 except tetrahydrofuan (THF) solvent was used.
  • THF tetrahydrofuan
  • Tyr.Me is a novel and effective initiator for the polymerization of amino acid NCA's.
  • Sample No. 2-13 represent a polymerization initiated with ⁇ -alanine and terminated wit: succinic anhydride.
  • ⁇ - aaanine is insoluble in most organic solvents, the reaction was carried out in refluxing THF.
  • the polydispersity of the polymer obtained was broader than that of the polymers initiated by Tyr.Me.
  • the molecular weight of the polymers was found to be dependent on the concentration of the monomer [M]. Low molecular weight polymers with broad distribution were obtaine ⁇ from a low [M] ( Figure 2, curve A). On the other hand, when [M] was greater than 0.2 g/mL, a polymer with a bimodal molecular weight distribution was obtained ( Figure 2, curve B). The lower molecular we it oligomers (-1000) may be due to an intramolecular termination between the terminal amino and the ⁇ -carboxylic groups.
  • the example illustrates a preferred method for the removal of benzyl protective groups in poly (Asp.Bz) and poly (Glu.Bz) by catalytic hydrogenation.
  • the hydrogenation of the polymers was carried out according to the following procedure: To a solution of the polymer in THF/methanol (1:1, v/v), Pd on active carbon (10%) was added in the amount of 1/10 of the polymer weight. After the replacement of air by nitrogen, hydrogen gas was introduced into the system and maintained with a balloon. The reaction mixture was stirred at room temperature overnight. After removing the catalyst by filtration and concentrating the solution, the mixture was poured into a large amount of petroleum ether to precipitate the polymer. The polymer obtai ed was then dried in vacuo.
  • This Example illustrates a method for the preparation and cleaning of empty proteinoid carriers.
  • This experiment describes encapsulation of anti-reovirus monoclonal antibody (mAb) 9BG5, an mAb directed against the sigma-1 gene product (Hemaglutinin, HA3) of the Reovirus Type
  • HA3 binds to the cell surface receptor for Reovirus type 3, and mAb 9GB5 interferes with viral binding to the receptor.
  • Mouse IgG monoclonal antibody 9BG5 was prepared and purified as described W.V. Williams et al. (1991) J. Biol. Chem., Vol. 266(8), pages 5182-5190, as well as references cited therein, using a purified Reovirus type 3 preparation (W.V. Williams et al. (1988) Proc. Natl. Acad. Sci. U.S.A, Vol. 85, pages 6488-6492).
  • the purified 9BG5 used in this Example had a protein concentration of 1.5 mg/ml in phosphate buffered saline (pH 7.2).
  • Proteinoid carriers encapsulating mAb 9BG5 were prepared having final concentrations of Glu/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Glu, Asp, Tyr, and Phe in the reaction mixture) 50 mg/ml, mAb 0.7 mg/ml and gum arable 0.5% in 0.85 N citric acid. Empty proteinoid carriers were prepared to contain the same final concentrations, except mAb was omitted. Aliquots (0.5 ml), in duplicate, of both mAb and empty proteinoid carriers preparations were centrifuged at 5000 RPM. Pellets and supernatants were frozen prior to analysis by Western blotting to determine antibody encapsulation efficiency.
  • Figure 6 is an x-ray film of a western blot analysis of purified mAb 9BG5, empty proteinoid carriers (no mAb added), and proteinoid carriers containing 9BG5. The analysis was done by immunoblotting with anti-mouse IgG which specifically reacted with mAb 9BG5.
  • the lanes correspond to the following:
  • the relative mobility (molecular weight) of the pure mAb is slightly different than the mAb in the proteinoid carriers. This is most likely due to different salt concentrations in the samples, because the encapsulation process employed 0.8 M salt solution.
  • the mAb 9BG5 preparations used to prepare the encapsulated proteinoid carriers had a protein concentration of approximately 2 mg/ml in phosphate buffered saline.
  • Final proteinoid concentration was 50 mg/ml and 5% (w/w) gum acacia ("gum”) or gelatin ("gel”). All proteinoid carriers were prepared in 0.85 N citric acid. Empty carriers were included for use as controls, and they were prepared in the same manner with the omission of mAb. Duplicate (0.5 ml) aliquots of proteinoid carrier suspension were centrifuged at 5000 RPM. Pellets and supernatants were frozen in dry ice prior to analysis.
  • Table 7 lists samples that were prepared. Numbers in parenthesis indicate amount of mAb added.
  • the samples were analyzed by conventional Western blotting as described in Example 9. Pellets were dissolved in sodium dodecyl sulfate with 0.05 N NaOH and analyzed under reducing conditions (breaks up the mAb into 50 kDa and 25 kDa bands). Aliquots (50 ⁇ l) of supernatants were analyzed under non- reducing conditions (expected intact 50 kDa mAb). This was done to determine differentially whether the mAb left behind is denatured or intact.
  • pellets of samples 9 and 10, and 11 and 12 contain between 5 and 10 ⁇ g of mAb.
  • the washed samples did not lose any significant amount of mAb, suggesting that the proteinoid carriers remained intact after freeze-thawing.
  • Sample 17 had some mAb encapsulated which was lost after washing (see number 18). This sphere preparation was not resistant to freeze-thawing. Additionally, a band at a MW of 150 kDa for sample 17 supernatants indicates that a significant amount of mAb is left behind after proteinoid carrier formation.
  • a mAb 9BG5 proteinoid carrier preparation and unencapsulated mAb 9BG5 were evaluated in rats.
  • the mAb proteinoid carriers suspension contained 0.25 mg/ml mAb and 50 mg/ml proteinoid in 0.85 N citric acid-0.5% gum. Empty proteinoid carriers were prepared similarly, but did not contain mAb.
  • the mAb proteinoid carriers were estimated to contain 0.075 mg/ml mAb and this value was used to determine dosages.
  • the mAb proteinoid carriers were examined microscopically and appear to be a fairly homogeneous preparation.
  • a purified mAb solution (0.95 mg/ml mAb in 0.85 N citric acid-0.5% gum) was used for oral gavage. This solution was prewarmed to 40oC prior to administration. For IV administration, a purified mAb solution (1 mg/ml mAb in phosphate buffer saline) was used.
  • mAb 9BG5 proteinoid carriers 3.7 mg mAb/ kg body weight of rat by oral gavage (rat # 2287, 2288, 2290, and 2291).
  • unencapsulated mAb 9GB5 0.73 mg/ kg body weight of rat by intravenous administration (rats #2292, 2293, and 2311).
  • unencapsulated mAb 9BG5 3.7 mg/ kg body weight of rat by oral gavage (rats #2314 and 2315).
  • Baseline blood samples (1 ml aliquots) were withdrawn from each rat just prior to dosing ("0" time). After dosing, blood samples were drawn at 1 h, 6 h and 24 h. The blood samples were processed immediately and sera were stored frozen at -20°C.
  • VLSH peptide (W.V. Williams et al (1991) J. Biol. Chem., Vol. 266(8), pages 5182-5190). Control plates included wells having no immobilized reovirus and V L SH peptides to which mAb (1mg/ml) was added.
  • VLSH peptide (W.V. Williams et al. ibid, Table 1) is a synthetic variant of VL peptide, the latter which corresponds to a portion of the light chain variable CDR II re on of 87.92.6 antibody.
  • the 87.92.6 antibody displays idiotypic and anti-idiotypic behavior towards reovirus type 3 receptor and mAb 9BG5, respectively (W.V. Williams et al. ibid).
  • the bound protein content of each well were measured by standard protein methods, e.g., Lowry method, and the results for each multi-well plate are shown in Figures 8(a-c), respectively.
  • Figures 8 (a-c) illustrate the levels of serum proteins which bound to immobilized reovirus type 3 and V L SH as detected by measurement protein concentration. These Figures show that the serum levels of bound proteins, after 24 hours post-dosing, were highest for animals orally administered mAb proteinoid carriers and animals administered unencapsulated mAb by the IV route. Lower levels of bound serum proteins were found in animals orally adminstered uncapsulated mAb. Serum taken from the animals receiving empty proteinoid carriers (no mAb) showed non-specific serum IgG protein binding, as expected, under the assay conditions.
  • Figure 9 show mAb binding under conventional ELISA procedures using immobilized reovirus type 3 and V L SH proteins.
  • Figure 9(a) or phosphate buffered saline ( Figure 9 (b) were employed.
  • the Figures show that the bound protein levels were higher for mAb in citrate buffer than for mAb in phosphate. Without being bound by any theory of operation for this invention, it is believed that the binding enhancement may be due to changes in the three dimensional conformation resulting from citrate-protein binding.
  • serum levels of mAb were greater in animals receiving encapsulated mAb by the oral route or unencapsulated mAb by the IV route, than an animal receiving orally administered unencapsulated mAb.
  • This Example describes a method for the preparation and cleaning of heparin proteinoid carriers.
  • Dialysis membrane tubing (Spectrum 6, 10 mm, 50,000 M.W. Cutoff)
  • proteinoid carrier encapsulates prepared with citric acid solutions are preferably dialyzed against 5% acetic acid solution for at least two hours with at least four changes of the dialysis solution to remove citric acid by an exchange process.
  • a Transfer the suspension with a syringe (no needle) to dialysis tubing and seal with plastic closures. Tubing should be no more than 70% full.
  • b Discard any amorphous material sedimented and/or aggregated on the surface.
  • c Dialyze the proteinoid carrier suspension against acetic acid solution (using 20 mL of acetic acid solution per ml of proteinoid carrier suspension) while stirring the acetic acid solution with a magnetic stirrer.
  • d Replace the acetic acid solution every hour. Continue dialyzing for a total of 3 hours. 6. Lyophilization:
  • a Weigh the lyophilized powder and calculate the amount of proteinoid in the powder.
  • b Add aqueous 0.85 N citric acid into the lyophilized powder at 40°C. The final concentration of proteinoid in solution is 80 mg/ml.
  • This Example illustrates a method for the preparation of insulin proteinoid carriers.
  • Proteinoid solution and insulin solution are combined at equal volumes sufficient to produce the final desired volume of proteinoid carriers.
  • Encapsulation of human erythropoietin (EPO) in proteinoid carriers was performed in the same manner described in
  • Rats weighing 50-200 grams are anesthetized with ketamine (8.5mg/kg) and thorazine 3.75mg/kg) with intramuscular injection.
  • the rat is then administered either unencapsulated erythropoietin or encapsulated erythropoietin by oral gavage.
  • an 8 french nelaton catheter is inserted down the esophagus of the rat until the 10cm mark on the catheter is even with the incisors.
  • the test or control solution is drawn up into a syringe and attached to the catheter. Holding the animal upright, the solution is expressed into the stomach of the rat.
  • the experimental results are summarized in Figures 10-12.
  • Serum erythropoietin levels were determined over time with an erythropoietin enzyme immunoassay kit (Amgen, Thousand Oaks, CA, USA). The results show that EPO serum levels in rats administered erythropoietin proteinoid carriers were relatively higher at all time points compared to rats (control) which received unencapsulated material.
  • the EPO levels remained at approximately 300 pg/mL serum in rats administered erythropoietin proteinoid carriers while the control rats had undetectable EPO levels.
  • Figure 11 illustrates EPO serum levels in rats that were administered either erythropoietin (50 ⁇ g/kg) or Gln/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Gln, Asp, Tyr, and Phe in the reaction mixture) proteinoid carrier encapsulated erythropoietin (50 ⁇ g/kg) directly into the proximal duodenum. Serum erythropoietin levels were determined over time with the aforementioned erythropoietin enzyme immunoassay kit.
  • Gln/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Gln, As
  • Calcitonin a peptide hormone which acts predominantly on bone to lower serum calcium concentration
  • Calcitonin proteinoid carriers were prepared by mixing a 1:1 volume ratio of a 100mg/ml aqueous solution of Gln/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Gln, Asp, Tyr, and Phe used in the proteinoid reaction mixture) and a 150 ug/mL calcitonin solution in 1.7 N citric acid solution with 1% gum acacia, as described in Example 13.
  • the efficiency of calcitonin encapsulation was approximately 40%.
  • Calcitonin concentration was determined directly by HPLC after dissolving the calcitonin proteinoid carriers in 60% aqueous acetonitrile.
  • the calcitonin proteinoid carriers prepared as described in Example 16, were evaluated in cynomolgus monkeys.
  • a single oral dose of calcitonin proteinoid carriers (0.25 mg/kg body weight) was administered to each of four monkeys by nasogastric gavage. The dosage was based on the body weight taken on the morning of dosing.
  • the hypocalcemic response following oral calcitonin administration was used as an index of pharmacological response.
  • Serum calcium concentrations were quantitated by a conventional O-cresolphthalein complexone method.
  • Figure 13 demonstrates the response obtained in cynomolgus monkeys following naso-gastric gavage of microencap sulated calcitonin. Significant changes from baseline serum calcium concentration were observed. Six hours following dosing, serum calcium concentrations decreased by 13 ⁇ g/ml. A significant pharmacological response was still apparent seven hours after the administration of calcitonin proteinoid carriers.
  • the calcitonin proteinoid carriers prepared in accordance with Example 16 are evaluated in fasted male Spraque Dawley rats weighing 100-150g. Calcitonin proteinoid carriers and calcitonin were administered by either oral gavage or intraduodenal injection. The rats are divided into the following groups:
  • calcitonin proteinoid carriers 60 ug calcitonin/kg body weight by oral gavage (3 rats);
  • calcitonin proteinoid carriers 3 ug calcitonin/kg body weight by intraduodenal gavage (3 rats);
  • calcitonin 3 ug calcitonin/kg body weight by intraduodenal gavage (3 rats) (Control).
  • Calcitonin proteinoid carriers are prepared immediately prior to dosing and Groups 1 and 2 each receive an appropriate dosage of the proteinoid carrier suspension. Groups 3 and 4 receive the unencapsulated calcitonin (no proteinoid carriers). Approximately 0.5 ml of blood is serially withdrawn from the tail artery of each rat just prior to dosing ("0" time) and 1 h, 2 h and 3 h post-dosing Serum from the blood samples are stored at -20°C for serum calcium concentration determination.
  • Figure 14 is the serum concentration-time curve for orally administered microencapsulated calcitonin and unencapsulated calcitonin in rats.
  • Experimental results in rats demonstrate a significant increase in pharmacological response (i.e., decreasing serum calcium levels) when proteinoid encapsulated calcitonin is compared to the unencapsulated vehicle control group.
  • serum calcium concentrations decreased 23 ⁇ g/ml in the rats receiving encapsulated calcitonin compared to a decrease of only 6.5 ⁇ g/ml in the control group.
  • the responses were dose-dependent (data not shown).
  • Example 17 The results obtained in this Example and in Example 17 provide evidence that proteinoid encapsulation markedly improves the oral bioavailability of calcitonin. The data also indicate that the oral drug delivery system is not species-dependent.
  • Factor IX is a vitamin K-dependent blood coagulation proenzyme, MW 56 kD.
  • Factor IX deficiency known as hemophilia B, occurs in approximately 1 out of every 25,000 males. To date, treatment of this disorder is accomplished by intravenous administration of Factor IX, although a recent report details efforts to supplement by subcutaneous injection (Thompson (1986) Blood, Vol. 67(3), pages 565-572).
  • FIX Factor IX
  • FIX proteinoid carrie ⁇ spension A contained 50 mg/ml of proteinoid and 500 U/ml FIX (FIX is available from the
  • the second suspension, FIX proteinoid carrier suspension B contained 50 mg/mi proteinoid and 116 U/ml FIX solution containing 3.8% acetic acid, 1.5% gum acacia, 0.15% PEG 14, 11 mM CaCl 2 , final pH 4.58.
  • FIX proteinoid carrier preparations The stability of FIX proteinoid carrier preparations was assessed over a sho: time course in vitro.
  • the protein carriers encapsulating FIX were examined by optical microscopy and laser light scattering. Aliquots of proteinoid carrier suspension were withdrawn every 30 minutes for 1.5 hours, FIX proteinoid carriers were isolated by centrifugation at 4500Xg and dissolved in activated partial thromboplastin time (APTT) assay buffer (0.05M histidine-0.01M NaCl-0.1% bovine serum albumin0.01% TWEEN-40, pH 7.47) to release soluble FIX and proteinoid.
  • APTT activated partial thromboplastin time
  • Quantitation of FIX activity by APTT employed both FIX standards (0.025, 0.05, and 0.1 U/ml) and "empty" proteinoid carrier suspension as control.
  • APTT assay kits are commercially available, e.g. Sigma Diagnostics (St. Louis, MO, USA).
  • FIX proteinoid carriers of greater stability are obtained by encapsulating FIX at a higher pH, e.g., pH 4.9. Furthermore, the efficiency of encapsulation is approximately 20% of available FIX units and activity levels remain constant for at least 1.5 hours when FIX proteinoid carrier pellets are stored at about 4oC.
  • FIX proteinoid carrier suspension A prepared as described in Example 19, were evaluated in male Sprague Dawley rats (ave. weight 300g). Appropriate aliquots of suspension were centrifuged at 4500Xg to pellet the FIX protein carriers, which were subsequently resuspended in the same buffer for animal dosing. The rats are divided into two groups as follows:
  • FIX proteinoid carriers 2709 U FIX/kg body weight by intragastric gavage (4 rats);
  • FIX IV Intravenous FIX (no proteinoid carriers)
  • FIX proteinoid carrier suspension and solution are prepared immediately prior to dosing.
  • One ml of blood was withdrawn from each rat just prior to dosing ("0" time) and 1 h, 2 h and 4 h (post-dosing), a citrate anticoagulant was added to the blood, and plasma from the blood samples were stored at -70°C.
  • Plasma samples were assayed by a modified APTT assay using FIX coagulated deficient plasma (assay kit is available from Ortho Diagnosis (Raritan, New Jersey, USA). Changes in clotting times were calculated by subtracting individual baseline (0 hr) values from subsequent clotting time values. The data shown in Figure 16 are the mean values for a given group. Values below baseline indicate the presence of exogenous FIX.
  • FIX proteinoid carrier suspension B prepared as described in Example 19, were evali ed in male Sprague Dawley rats (avenuit weight 300g). Resuspended FIX proteinoid carriers were prepared as described in Example 20.
  • the rats are divi led into two groups as follows:
  • FIX proteinoid carriers 1006U FIX/kg body weight by intragastric gavage (5 rats).
  • FIX IV Intravenous FIX (no proteinoid carriers)
  • 3 rats received 0.3 ml FIX in 0.11 NaCl-0.02M sodium citrate, pH 6.85 by tail vein injection.
  • FIX no proteinoid carriers
  • FIX unencap PO 2760U FIX/kg body weight by intragastric gavage. 4 rats received
  • FIX proteinoid carrier suspension and solutions were prepared immediately prior to dosing. Plasma samples were obtained and assayed as described in Example 20. Changes in clotting times were calculated by subtracting individual baseline
  • FIX proteinoid carriers support that oral delivery of FIX can be accomplished via the use of FIX proteinoid carriers. These proteinoid carriers appear to adequately protect FIX during transit through the GI tract and deliver FIX to the blood stream.
  • IFN alpha-Interferon
  • IFN proteinoid carriers were prepared with an aqueous solution of Glu/Asp/Tyr/-Phe proteinoid (1:1:1:1 mole ratio of Glu, Asp, Tyr and Phe used in the proteinoid reaction mixture), and an IFN solution containing 1.7 N citric acid solution with 5% gelatin.
  • the IFN proteinoid carrier suspension contained 80 mg/ml proteinoid, 600 ug/ml IFN, 0.6N citric acid, and 2.5% gelatin, pH 3.0. Stability of IFN proteinoid carriers in SGF
  • IFN proteinoid carriers were much more stable than IFN alone (in the absence of proteinoid) m SIF.
  • IFN alone at pH 7.4 was completely degraded within 10 minutes when incubated with SIF.
  • proteinoid carriers are required for protective capability or whether (1) proteinoids (soluble proteinoids- -not in carrier form) may be used and whether (2) alternative methods of carrier loading, such as incubating the therapeutic compound with preformed proteinoid carriers, are useful.
  • Heparin proteinoid carriers were prepared, following the procedure of Example 12, using a 1:1 volume ratio of 150 mg/ml of Glu/Asp/Tyr/Phe/Orn 0.5 (1:1:1:1:0.5 mole ratio of Glu, Asp, Tyr, Phe, and Orn used in the proteinoid reaction mixture) proteinoid in deionized water, and an 20mg/mL aqueous heparin solution containing 1.7 N citric acid solution and 0.5% gum acacia.
  • the heparin proteinoid carrier suspension was dialyzed in acetic acid solution as described in Example 12.
  • Heparin proteinoid carriers were then centrifuged at 4800Xg (15 minutes) and total heparin was measured by assaying the pellet and the supernatant with a modification of the Azure A method (Gundry et al. Amer. J. of Surgery (1984) Vol. 148, pages 191-194). Proteinoid was assayed by dissolving the proteinoid carriers with 0.1 N NaOH and measuring absorbance at 294 nm.
  • Empty proteinoid carriers were prepared following the same procedure described above for the heparin proteinoid carriers, with the modification being that no heparin was present.
  • the lyophilized empty proteinoid carriers were resuspended in 0.85N citric acid and 0.5% gum containing heparin at a concentration of 20 mg/ml.
  • the amount of heparin coisolated with the proteinoid carriers was measured as described above.
  • Rats Male Spaque Dawley rats weighing approximately 350g were dosed by oral gavage or intraduodenal (ID) injection (just anterior to the pyloric sphincter and into the duodenum). Rats were dosed orally or ID with one of the following: lyophilized heparin proteinoid carriers, heparin-spiked empty proteinoid carriers, proteinoid/heparin in water, heparin in 0.85N citric acid and 0.5% gum and heparin alone in water. In both oral and ID injection experiments, weight ratios of neparin:proteinoid were constant. The total heparin dose in the oral studies was 100 mg/kg body weight; in ID injections studies, it was 50 mg/kg.
  • ID intraduodenal
  • the proteinoid dose was 40 mg/kg for oral gavages and 20 mg/kg for ID injections.
  • the dosing volume was approximately 0.3 to 0.5 ml.
  • Approximately 0.5 ml of blood is serially withdrawn from the tail artery of each rat just prior to dosing ("0" time) and 1 h, 2 h and 4 h post-dosing. Serum from the blood samples are stored at -20°C for heparin activity determination.
  • Heparin proteinoid carriers gave the highest APTT values, indicated increased absorption of heparin when dosed orally, as well as when directly injected into the duodenum
  • Proteinoid carrier In this Example, influenza virus antigen-containing proteinoid carriers were prepared and evaluated in rats.
  • M1 protein a major internal component of influenza virus, was obtained by purification of a swine influenza vaccine donated by Drug Directorate, Health Protection Branch, Bureau of Biologies, Ottawa, Ontrario Cannada.
  • the vaccine was prepared with the high-yielding recombinant strain X-53Aa, which derives its HA and NA from the parent strain A/NJ/11/76 (H1N1) and its internal proteins, including M1, from the parent strain A/PR/8/34 (R.B. Couc et al. (1983) Ann. Rev. Microbiol., Vol. 37, pages 529-549 and B.R. Murphy (1982) Infec.
  • M1 was purified as described by Khan et al ( (1982) J.Clin.Microbiol., Vol. 16, pages 813-820).
  • M1 proteinoid carriers were prepared, by mixing (at 40°C), equivolumes of an aqueous solution of 100mg/ml of Glu/Asp/Tyr/Phe proteinoid in deionized water and a 10mg/mL solution of M1 protein in 1.7N citric acid and 5% gum arabic (pH 2.0). The final M1 concentration in the suspension was 1.0mg/ml.
  • HA-NA antiger was isolated according to the procedure of Gallagher et al. ((1984) J. Clin.Microbiol., Vol. 20, pages 80-93). Influenza virus (A/PR8/34) was centrifuged at 90,000 G for 60 min. The viral pellets was solubilized with 0.05M acetate buffer (pH 7.0) containing 7.5% octylglucoside and re-centrifuged under the same conditions. The resulting supernatant contained approximately 90% HA and 10% NA as determined by SDS-PAGE.
  • HA-NA proteinoid carriers were prepared following the same protocol as for the M1 proteinoid carriers but substituted M1 for HA-NA.
  • the final concentration of HA-NA in the suspension was also 1.0 mg/ml.
  • mice Male Spraque Dawley rats (about 350g weight) were used in this experiment. Oral dosage was by gavage.
  • SC subcutaneously
  • Serum anti-M1 and anti-HA-NA specific IgGs were assayed by an ELISA method as described Khan et al. ((1982) J. Clin. Microbiol., vol. 16, pages 813-820).
  • Plasma samples from rats dosed orally with "empty" proteinoid carriers showed no significant antibody titer against either M1 or HA-NA antigens when assayed by ELISA (Table 8).
  • rats dosed with 25 ug of either M1 or HA-NA antigen showed no significant antibody titer against either M1 or HA-NA antigens when assayed by ELISA (Table 8).
  • Plasma samples from three of the five rats dosed with M1 proteinoid carriers showed a signficant primary response to M1 antigen. All three rats had titers ranging from 760 to 2150 as early as 14 days post-dosing, compared to ⁇ 30 in all rats that received the amount of unencapsulated M1 (Table 8). Titers in the group that received proteinoid carriers increased to 1150-5200 by 42 days ( Figure 24).
  • PA phosphoric acid
  • Equ equxients
  • GLYC glycerol
  • PPA polyphosphoric acid

Abstract

Improved proteinoid carriers and methods for their preparation and use as oral delivery systems for pharmaceutical agents are described. The proteinoid carriers are soluble within selected pH ranges within the gastrointestinal tract and display enhanced stability towards at least one of photolysis or decomposition over time. The proteinoid carriers are prepared from proteinoids having between 2 and 20 amino acids and having a molecular weight of between about 250 and 2400 daltons.

Description

PROTEINOID CARRIERS AND METHODS
FOR PREPARATION AND USE THEREOF
This application is a continuation-in-part of U.S. applicr on serial no. 07/920,346, filed July 27, 1992, which in turn is a continuation-in-part of U.S. application serial no. 07/898,909, filed June 15, 1992.
Field of the Invention
This invention relates to proteinoids and proteinoid carriers made from them. The proteinoid carriers releasably encapsulate active agents and have extended longer shelf life and/or photostability. Methods for the preparation of such proteinoid carriers are also disclosed. Background of the Invention
The available modes of delivery of pharmaceutical and therapeutic agents often are severely limited by chemical or physical barriers or both, which are imposed by the body. For example, oral delivery of many such agents would be the route of choice if not for the presence of chemical and physicochemical barriers such as extreme pH in the gut, exposure to powerful digestive enzymes, and impermeability of gastrointestinal membranes to the active ingredient. Among the numerous pharmacological agents which are Known to be unsuitable for oral administration are biologically active pepuides and proteins, such as insulin. These agents are rapidly destroyed in the gut by acid hydrolysis and/ or Joy proteolytic enzymes.
A great deal of research has been devoted to developing effective oral drug delivery methods and systems for these vulnerable pharmacological agents. The proposed solutions have included :
(a) co-administration of adjuvants (such as resorcinols and non- ionic surfactants polyoxyethylene oleyl ether and n-hexadecyl polyethylene ether to increase the permeability of the intestinal walls; and
(b) co- administration of enzymatic inhibitors, such as pancreatic trypsin inhibitor, diisopropylfluorophosphate (DFF) and nrasylol to avoid enzymatic degradation.
The use of such substances, in drug delivery systems, is limited however either ioecause of:
(a) their inherent toxicicy when employed at effective amounts;
(b) their failure to protect the active ingredient or promote its absorption;
(c) their adverse interaction with the drug.
Liposomes as drug delivery systems have also been described. They provide a layer of lipid around the encapsulated pharmacological agent. The use of liposomes containing heparin is disclosed in U.S. Patent No. 4,239,754 and several studies have been directed co the use of liposomes containing insulin; e.g., Patel et al. (1976) FEBS Letters Vol. 62, page 60 and Hashimoto et al. (1979) Endocrinol. Japan, Vol. 26, page 337. The use of liposomes, however, is still in the development stage and there are continuing problems, including:
(a) poor stability;
(b) inadequate shelf life;
(c) limited to low MW (< 30,000) cargoes;
(d) difficulty in manufacturing;
(e) adverse interactions with cargoes.
More recently, synthetic amino acid polymers or proteinoids, forming microspheres, have been described for encapsulating pharmaceuticals. For example, U.S. Patent No. 4,925,673 (the '673 patent), the disclosure which is hereby incorporated by reference in its entirety, describes such microsphere constructs ai-well as methods for their preparation and use. The '673 patent also describe: microspheres which encapsulate pharmaceuti-cal agents for delivery into the gastrointestinal tract or into the blood.
While the proteinoid microspheres described in the '673 patent are useful for their intended purposes, the physicochemical properties of the proteinoid microspheres, such as light sensitivity, shelf life and the selectivity of their solubility in various portions of the gastrointestinal tract, could be improved. Additionally, there is a need in the art for microspheres that can encapsulate a broader range of active agents such as polar drugs.
The method employed in the '673 patent to prepare proteinoids produces a complex mixture of high molecular weight
(MW) (> 1000 daltons) and low MW (≤ 1000 daltons) peptide-like polymers which are difficult to separate. Moreover, the method produces a small amount of the low MW proteinoids which is the microsphere-forming fraction. Hence, an improved method of preparing of the proteinoids is also desired.
Accordingly, there is a need in the art for improved proteinoid carriers as well as improved methods for their preparation.
Objects of the Invention
It is an object of this invention to provide proteinoids which forms proteinoid carriers as a delivery system with enhanced stability towards at least one of photodegradation and decomposition over time.
It is anot er object of the invention to provide a proteinoid that forms proteinoid carriers with more selective solubility under various conditions such as pH.
It is yet another object of the invention to provide proteinoid carriers encapsulating biologically active agents which are selectively releasable within particular portions of the gastrointestinal tract.
It is a further object of the invention to provide proteinoid carriers which promotes the bioavailability of pharmaceutical agents which otherwise display poor absorption in the gastrointestinal tract.
It is yet a further object of the invention to provide an improved method for producing proteinoid carriers having particular characteristics and for improving yield of the desired proteinoid carriers .
It has been found that these objects and other advantages, which will be apparent from this specification, are achieved by the invention described below.
Summary of the Invention
The present invention relates to improved proteinoid carriers and methods of making and use thereof.
Proteinoids of a MW ranging between about 250 and about 2400 daltons and of defined amino acids are useful in preparing proteinoid carriers with improved stability against photodegradation and/or decomposition. The proteinoids comprise a peptide polymer selected from the group consisting of:
(i) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid;
(ii) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and from at least one third monomer selected from the group consisting of lysine, arginine and ornithine, the proteinoid being a microsphere- and/or microcap- sule-forming proteinoid and being soluble within a selected pH range.
The proteinoid molecules of the invention contain between about 2 and about 20 amino acid residues, preferably between about 2 and about 8 amino acid residues, and has a molecular weight which ranges between about 250 and about 2400 daltons, preferably between about 250 and about 600, and most preferably between about 250 and 400 daltons.
The proteinoid carriers are useful as delivery systems to releasably encapsulate and carry a broad range of cargoes including pharmaceutical agents, dye reagents and cosmetic ingredients. In particular, the proteinoid carriers are useful as oral delivery systems of sensitive pharmaceutical agents, which normally would not be admmistrable via the oral roυte for selective release at targeted regions of the gastrointestinal tract.
Description of the Drawings
Figure 1 illustrates the molecular weight distribution as a function of monomer concentration of poly (Asp.Bz-co-Phe) polymer prepared by the NCA method as described in Example 3.
Figure 2 illustrates the molecular weight distribution of a function of monomer concentration of poly (Asp.Bz) polymer prepared by the DPPA method as described in Example 5.
Figure 3 illustrates the effect of reaction time duration on yields of poly (Asp.Bz) polymer prepared by the DPPA method as described in Example 5.
Figure 4 illustrates be effect of temperature of the molecular weight of poly (Asp.Bz) polymer prepared by the DPPA method as described in Example 5.
Figure 5 illustrates the effect of changing the molar ratios of [DPPAl/ [M] on the molecular weight of poly (Asp.Bz) polymer by the DPPA method as described in Example 5.
Figure 6 is a photograph of an x-ray film of the western immunoblot analysis, as described in Example 9, of purified murine mAb 9BG5 (2μg, lane 1; lmg, lane 2; and 0.25 μg, lane 3); empty proteinoid carrier supernatant after encapsulating process (no mAb) (lane 4); empty proteinoid carrier pellet (lane 5); proteinoid carrier encapsulated mAb supernatant after encapsulating process (lane 6); and proteinoid carrier encapsulated mAb pellet. Lane MW contained standard molecular weight markers.
Figure 7 is a photograph of an x-ray film of a western immunoblot analysis of samples described in Example 10.
Figures 8 (a-c) illustrate the levels of serum proteins which bound to immobilized reovirus type 3 and VLSH under ELISA conditions as described in Example 11. "Empty spheres" refers to animals orally administered empty proteinoid carriers (no mAb 9BG5); "mAb spheres" refers to animals orally administered mAb 9BG5 encapsulated proteinoid carriers; "IV" refers to animals intravenously administered unencapsulated mAb 9BG5; and "oral" refers to animals orally administered unencapsulated mAb 9BG5.
Figure 9 show mAb binding under conventional ELISA procedures using immobilized reovirus type 3 and VLSH proteins with serial dilutions of purified mAb in 0.85 N citrate-0.5% gum (Figure 9 (a)) or phosphate buffered saline (Figure 9 (b)) as described in Example 11.
Figure 10 illustrates levels of erythropoietin (EPO) detected in rat serum taken from rats administered proteinoid carrier encapsulated EPO (15μg EPO/kg body weight) and encapsulated EPO (15μg EPO/kg body weight) as described in Example 15.
Figure 11 illustrates EPO serum levels in rats that were administered either erythropoietin (50μg/kg) or encapsulated erythropoietin (50μg/kg) directly into the proximal duodenum as described in Example 15. Serum erythropoietin levels were determined over time with a erythropoietin enzyme immunoassay kit.
Figure 12 illustrates EPO serum levels in rats who were orally gavaged with either encapsulated or unencapsulated erythropoietin (100μg/kg) or received a subcutaneous injection of either 2μg/kg or 10μg/kg as described in Example 15. Serum erythropoietin levels were determined over time with an erythropoietin enzyme immunoassay kit.
Figure 13 illustrates serum calcium changes after oral administration of salmon calcitonin proteinoid carriers (0.25 mg calcitonin/kg body weight) in cynomolgus monkeys as described in Example 17. The results are expressed as absolute change in serum calcium from baseline values. The data represents means +/- SEM. ** Serum calcium levels significally different from baseline values.
Figure 14 illustrates serum calcium changes following oral administration of salmon calcitonin proteinoid carriers (0.60 mg/kg body weight) in rats as described in Example 18. The results are expressed as absolute change in serum calcium from baseline values. The data represents means +/- SEM. **Serum calcium levels significantly different compared to the control group at the corresponding time point.
Figure 15 illustrate, serum calcium changes after intraduodenal administration of salmon calcitonin or calcitonin proteinoid carriers (3 ug/kg body weight) in rats as described in Example 18. The results are expressed as absolute change in serum calcium from baseline values. The data represents means +/- SEM. ** Significantly different from the unencapsulated control group at the indicated time points.
Figure 16 illustrates clotting times after oral administration of proteinoid carrier encapsulated Factor IX (FIX sph PO) and IV administration of FIX solution (FIX IV) as described in Example 20.
Figure 17 illustrates clotting times after oral administration of proteinoid carrier encapsulated Factor IX (FIX sph PO) and FIX solution (FIX unencap PO) or IV administration of FIX solution (FIX IV) as described in Example 21.
Figure 18 illustrates the percentage of intact alpha- interferon (IFN) remaining after incubating IFN and IFN proteinoid carriers in simulated gastric fluid (SGF). Figure 19 illustrates the percentage of intact IFN remaining after incubating IFN and IFN proteinoid carriers in 0.08N HCl.
Figure 20 illustrates the percentage of intact IFN remaining after incubating IFN and IFN proteinoid carriers in simulated intestinal fluid (SIF).
Figure 21 illustrates the clotting times in rats dosed with heparin or proteinoid/heparin, both in water. The data represents an average of 6 rats. The data represents means +/- SEM.
Figure 22 illustrates clotting times in rats dosed ID with USP heparin or heparin proteinoid carriers, both in citric acid. Each time point is an average of 12 rats. The data represents means +/- SEM.
Figure 23 illustrates clotting times in rats dosed orally with heparin-spiked empty proteinoid carriers or heparin proteinoid carriers. Each time point is an average of 12 rats. The data represents means +/- SEM.
Figure 24 illustrates the average titers of rats immunized orally with M1 proteinoid carriers versus unencapsulated M1. Only responders in each group were averaged.
Figure 25 illustrates HA-NA titers of rats immunized orally with HA-NA micropspheres versus unencapsulated HA-NA. Detailed Description of the Invention
All patents and literature references cited in this specification are hereby incorporated by reference in their entirety. In case of inconsistencies, the present description, including the definitions and interpretations, will prevail.
The instant invention arose from the discovery that proteinoids of a MW of between about 250 and about 2400 daltons and of defined amino acid composition can be obtained by modifying known reactions and selecting starting materials. These proteinoids form proteinoid carriers with surprisingly enhanced stability against at least one of photodegradation and decomposition over time. In addition, proteinoid carriers prepared from such proteinoids carr. a broader range of pharmaceutical agents, including labile polypeptides such as insulin, alpha-interferon, calcitonin, antigens, e.g. influenza virus M1-protein, and Factor IX and display a selective releasability within various portions of the gastrointestinal tract, relative to prior art proteinoid microspheres.
The proteinoids of the invention comprise a peptide polymer selected from the group consisting of:
(i) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid;
(ii) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and from at least one third monomer selected from the group consisting of lysine, arginine and ornithine, the proteinoid being a microsphere- or microcapsule-forming proteinoid and being soluble within a selected pH range.
The proteinoid molecules of the invention contain between about 2 and about 20 amino acid residues, preferably between about 2 and about 8 amino acid residues, and have a molecular weight which ranges between 250 and about 2400 daltons, preferably between about 250 and about 600, and most preferably between about 250 and 400 daltons.
Proteinoid carriers prepared from the proteinoid molecules, in accordance with the present invention, display a selective solubility at specific acidic or basic pH ranges, depending on the choice and amount of the second and third monomers in the proteinoid.
Proteinoid carriers which are selectively soluble under alkaline pH environments sue: as those found in the distal portion of the intestine, are prepared from base-soluble proteinoids. These proteinoids contain, as starting monomers in the reaction mixture, at least one second monomer selected from the group consisting of glutamic acid, glutamine, pyroglutamic acid, and aspartic acid. At a pH ranging between about 7.2 and about 11.0, the base-soluble proteinoid exists largely as the anion and is soluble. At a pH below about 7.0, the proteinoid is largely protonated and insoluble in water.
Similarly, proteinoid carriers which are selectively soluble under acidic pH environments, such as the stomach, are prepared from acid-soluble proteinoids. In this case, the proteinoid contain, as starting monomers in the proteinoid reaction mixture, at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid and at least one third monomer selected from the group consisting of lysine, arginine, and ornithine. At a pH ranging between about 1 and about 7, the base-soluble proteinoid exists largely as the cation and is soluble. At a pH above about 7.2, the proteinoid is largely unprotonated and insoluble in water.
The pH and the solubility characteristics of the acid-soluble proteinoid depends largely, but not exclusively, upon the pH and solubilty of the last amino acid added during the synthesis of the proteinoid. For instance, the incorporation of a basic amino acid, e.g., a third monomer, selected from the group consisting of lysine, arginine and ornithine in the acid-soluble proteinoid will result in the elevation of the pI (pH at the isoelectric point) of the proteinoid.
The proteinoids of the present invention are preparable by a thermal condensation reaction by heating mixtures of the appropriate amino acids under conditions described in the '673 patent. In contrast with the '673 patent procedures which use as many as eighteen amino acids, mixtures of two to five specific amino acids with at least one selected from each of the aforementioned groups yield proteinoids which form proteinoid carriers with selective solubility at particular pH ranges and at nigh yields.
In carrying out the thermal condensation reaction, it has now been discovered that inclusion of tetramethylene sulfone, an inert, high boiling, polar solvent, maximizes the yield (> 80%) of low MW proteinoids. Omission of solvent does not produce high yields of low MW proteinoids. Presumably this is due to the poor solubility of the amino acid monomers in these solvents and/or unavoidable side reactions between the monomers and the solvent under the reaction conditions.
In general, individual amino acids are added to a reaction flask containing tetramethyl the sulfone (sulfolane) wnich has been heated to a temperatur ranging bet en about 130°C and about 200°C, preferably about 175°C to 195°C under an inert atmosphere of argon or nitrogen gas. After each addition, the solution is stirred for a period of time ranging between about 10 minutes id about hours, depending on the amino acid type and the order of addition.
Upon heating mixtures of amino acids to temperatures of about 190°C as described above, a reaction takes place and water, ammonia and carbon dioxide are produced as side-products. Water is removed from the reaction as formed and the reaction is terminated when water formation ceases. Thereafter, the proteinoid are precipitated out of the reaction solution by quenching with excess water, under vigorous stirring. After stirring for a period of about 1 hour, the proteinoids are collected by filtration, washed with water and dried under vacuum.
Chemical condensation methods which utilize derivatized amino acids are also useful for making the proteinoids of the present invention as they permit greater control of molecular weight. Such reactions are generally conducted at lower reaction temperatur and with initiators. In particular, low MW proteinoids produced by the alpha-amino acid N-carboxyanhydride (NCA) method and the diphenylphosphoryl azide (DPPA) method (N. Nishi et al. (1991) Makromol. Chem., Vol.192, pages 1789-1798) were found to form proteinoid carriers having selected solubility within a particular pH range.
The NCA method involves the preparation of N-carboxyanhydrides of alpha-amino acid esters and their subsequent polymerization, using low MW amines as initiators. It has been discovered that non-NCA derived amino esters, e.g., α-methyl tyrosine ester, are effective initiators which are stable and soluble in many organic solvents such as tetrahydrofuran (THF). The use of amino acids as initiators, presumably due to their poor solubility in organic solvents and their low stability, are not known. The NCA reaction produces a high yield of proteinoids with high purity.
The DPPA method involves the direct condensation of benzyl esters of alpha-amino acids in the presence of DPPA and a low MW amine, followed by removal of the protective benzyl groups, contained in the proteinoid product, by alkaline hydrolysis. If catalytic hydrogenation is used in place of alkaline hydrolysis, low MW proteinoids of unexpected high purities and yields are obtained.
Proteinoids prepared by any of the above methods can be used immediately to microencapsulate an active pharmacological agent or the proteinoid can be concentrated or dried by conventional means and stored for future use.
The proteinoids of the invention are purified as follows: crude proteinoids are slurried with water at room temperature, e.g. 25°C. While at this temperature, the pH of the slurry is adjusted to about pH 8 using an aqueous alkaline solution, e.g. 40% sodium hydroxide and 10% sodium bicarbonate solutions for an acid-soluble proteinoid. For a base-soluble proteinoid, the slurry is adjusted to an acidic pH with an aqueous acidic solution, e.g. 10% acetic acid solution. The mixture is then filtered and the filter cake washed with a volume of water. The washes and filtrate are then combined and evaporated to dryness in vacuo to afford proteinoids. If necessary, this process can be repeated until proteinoids of a desired purity level are obtained.
If desired, the proteinoid may be further purified by fractionating on a column containing solid supports which include silica gel or alumina, using methanol or propanol as mobile phase; ion exchange resin using water as the mobile phase; reverse phase column supports using trifluoroacetic acid/acetonitrile mixtures as mobile phase. The proteinoids may also be purified by extraction with a lower alcohol such as propanol or butanol to remove low molecular weight contaminants.
Proteinoid carriers are made from purified proteinoids as follows: proteinoids are dissolved in deionized water at a concentration ranging between about 75 and about 200 mg/ml, preferably about 100 mg/ml, at a temperature between about 25ºC and about 60ºC, preferably about 40°C. Particulates remaining in the solution may be filtered out by conventional means such as gravity filtration over filter paper.
Thereafter, the proteinoid solution, maintained at a temperature of about 40°C, is mixed with an aqueous acid solution (also at about 40°C) having an acid concentration ranging between about 1 N and about 2 N, preferably about 1.7 N. The resulting mixture is further incubated at 40°C for a period of time effective for microsphere and microcapsule formation as observed by light microscopy. In practicing this invention, the preferred order of addition is adding the proteinoid solution to the aqueous acid solution.
Suitable acids include any acid which does not (a) adversely effect the proteinoid, e.g., chemical decomposition;
(b) interfere with microsphere or microcapsule formation; (c) interfere with microsphere or microcapsule encapsulation of cargo; and (d) adversely interact with the cargo. Preferred acids for use in this invention include acetic acid, citric acid, hydrochloric acid, phosphoric acid, malic acid and maleic acid.
In practicing the invention, a proteinoid carrier stabilizing additives are preferably incorporated into the aqueous acid solution or into the proteinoid solution, prior to the microsphere or microcapsule formation process. The presence of such additives promotes the stability and dispersibility of the proteinoid carriers in solution.
The additives may be employed at a concentration ranging between about 0.1 and 5 % (W/V), preferably about 0.5 % (W/V). Suitable, but non-limiting, examples of stabilizing additives include gum acacia, gelatin, polyethylene glycol, and polylysine.
Thereafter, the proteinoid carriers may be used immediately or may be stored at 4ºC or lyophilized and stored under desiccant at room temperature or below.
Under the aforementioned conditions, the proteinoid molecules form spherical proteinoid carriers comprising proteinoid microcapsules and proteinoid microspheres of less than 10 micron diameter. As defined herein, a-"microsphere" is spherical homogeneous mesh work structure having no discrete inner chamber. A "microcapsule" refers to a spherical structure having a proteinoid wall which forms a hollow or chamber. If the proteinoid carriers are formed in the presence of a soluble material, e.g., a pharmaceutical agent in the aforementioned aqueous acid solution, this material is believed to be encapsulated within the hollows of the microcapsules and confined within the proteinoid wall defined by the spherical structure or entrapped within the matrix of proteinoid molecules in the microsphere structure. In this way, one can encapsulate or entrap pharmacologically active materials such as peptides, proteins, and polysaccharides as well as charged organic molecules, e.g., quinolones or antimicrobial agents, having poor bioavailability by the oral route. The amount of pharmaceutical agent which may be encapsulated or entrapped by the proteinoid carrier is dependent on a number of factors which include the concentration of agent in the encapsulating solution.
The proteinoid carriers of the invention are pharmacologically harmless and do not alter the physiological and biological properties of the active agent. Furthermore, the encapsulation pre ess does not alter the pharmacological properties of the active agent. While any suitable pharmacological agent can be encapsulated within proteinoid carriers, it is particularly valuable for delivering agents which otherwise would be destroyed or rendered less effective by conditions encourtered in the animal body before it reaches its target zone and which are poorly absorbed in the gastrointestinal tract.
The proteinoid carriers of the invention are particularly useful for the oral administration of certain pharmacological agents, e.g., small peptide hormones, swhich, by themselves, pass slowly or not at all through the gastro-intestinal mucosa and/or are. susceptible to chemical cleavaσe by acids and enzymes in the gastrointestinal tract. Non-limiting examples of such agents include human or bovine growth hormone, interferon and interleukin-II, calcitonin, atrial naturetic factor, antigens, monoclonal antibodies, and Factor IX,. a vitamin K-dependent blood coagulation proenzyme.
The choice of a particular proteinoid for use in encapsulating or entrapping a pharmacological agent depends on a number of factors which include:
(1) the acidity or basicity of the agent;
(2) the targeted area for release in the gastrointestinal tract;
(3) the solubility of the drug at certain pH ranges;
(4) efficiency of encapsulation;
(5) interaction of drug with proteinoid.
For example, proteinoids made from gl; tamic acid, aspartic acid, tyrosine, and phenylalanine are especially suitable for encapsulating polysaccharides like heparin.
In addit i on to selective pH solubility, the particle size of the proteinoid carrier plays an important role in determining release of the active agent in the targeted area of the gastrointestinal tract. Proteinoid carriers having diameters between about ≤ 0.1 microns and about 10 microns, preferably between about 5.0 microns and about 0.1 microns, and containing encapsulated or entrapped active agents are sufficiently small to effectively release the active agent at the targeted area within the gastrointestinal tract. Large proteinoid carriers (>10 microns) tend to be less effective as oral delivery systems.
The size of the proteinoid carriers formed by contacting proteinoids with water or aqueous solution containing active agents can be controlled by manipulating a variety of physical or chemical parameters, such as the pH, osmolarity or salt content of the encapsulating solution, and the choice of acid used in the encapsulating process.
By tailoring both the solubility characteristics of a proteinoid and the particle size of the proteinoid carriers, active agent bearing proteinoid carriers can be produced from base-soluble proteinoids which are stable in the highly acidic stomach (normal pH of from about 2 to about 6), but which dissolve in the distal portion of the intestines. Such systems are suitable for oral administration of peptide hormones, e.g., insulin, and polysaccharides, e.g., heparin, which otherwise would be quickly destroyed in the GI tract. They also are suitable for protecting the stomach from gastric irritants, such as aspirin. When such aspirin-containing proteinoid carriers are orally administered, they pass through the gastrointestinal mucosa and release the aspirin far more rapidly than conventional enterically coated aspirin, which first must traverse the stomach and then must enter the bloodstream from the intestine after the enteric coating has dissolved.
It also is possible to produce systems from acid-soluble proteinoids which are stable under weakly basic conditions (pH of about 8), but which release active agent under acidic conditions (pH of about 2 to 5). Such systems are suitable for the intravenous administration of pharmacological agents such as calcium regulators and redox carrier systems for dopamine or gamma-aminobutyric acid.
The proteinoid carriers of the invention may be orally administered alone as solids in the form of tablets, pellets, capsules, and granulates suitable for suspension in liquids such as edible oils. Similarly, the proteinoid carriers can be formulated into an orally administrable composition containing one or more physiologically ..ompatible carriers or excipients. These compositions may contain conventional ingredients such as gelatin, polyvinylpyrrolidone and fillers such as starch and methyl cellulose.
The proteinoid carriers of the invention may also be administered by injection.
The following examples are illustrative of the invention but are not intended to limit the scope of the invention. Example 1: Preparation of a Base-soluble Proteinoid
by a Thermal condensation Reaction
750 ml of tetramethylene sulfone was heated to 190°C in an inert nitrogen atmosphere in a 4 liter flask with stirring.
294 g of glutamic acid was added and the mixture was heated for one-half hour. 266 g of aspartic acid was added and the mixture heated as rapidly as possible to 190°C and held there for 15 minutes. 362 g of tyrosine was added and the mixture heated at 190°C for 3 hours. 330 g of phenylalanine was added and the mixture heated at 190°C for 1.5 hours. The hot melt was then poured into 5 liters of water with vigorous stirring. After stirring for about 1 hour, the mixture was filtered and the filtrate discarded. The cake was reslurried in 5 liters of water, filtered and the jake was again reslurried in 5 liters of water. The pH of the slurry (at 25°C) was adjusted to 8 using 40% sodium hydroxide solution. The mixture was filtered and the cake washed with a small amount of water. The washes and filtrate are combined and evaporated to dryness in vacuo to give Glu/Asp/Tyr/Phe proteinoid.
Appendices A, B, and C describe examples of other proteinoids prepared by the thermocondensation method. Example 2: Preparation of an Acid-soluble Proteinoid
by a Thermal Condensation Reaction
750 ml of tetramethylene sulfone is heated to 190°C in an inert nitrogen atmosphere in a 4 liter flask with stirring. 294 g of glutamic acid is added and the mixture is heated for one-half hour. 362 g of tyrosine is added and the mixture is heated at 190°C for 3 hours. 330 g of phenylalanine is added and the mixture is heated at 190°C for 1.5 hours. 266 g of arginine is added and the mixture is heated for an additional 1.5 hours. The hot melt is then poured into 5 liters of water with vigorous stirring. After stirring for about 1 hour, the mixture is filtered and the filtrate is discarded. The cake is reslurried in 5 liters of water, filtered and the cake is again reslurried in 5 liters of water. The pH of the slurry (at 25°C) was adjusted to 5 using 10% acetic acid solution. The mixture is filtered and the cake is washed with a small amount of water.
The washes and filtrate are combined and evaporated todryness in vacuo to give proteinoid.
Appendices A, B, and C describe examples of other proteinoids prepared by the thermocondensation method.
Example 3 : Preparation of Proteinoids by the
NCA Method Using Amine Initiator
This example illustrates the NCA method for preparing copolypeptides consisting of Asp.Bz, Glu.Bz, Phe, and Tyr components. The NCA monomers of these amino acids were prepared according to the reported method.
The reactions were carried out in tetrahydrofuran (THF) or in dichloromethane using benzylamine (BzNH2) or 4-methylbenzyl amine (MeBzNH2) as initiator at room temperature ( [M] = 10%). The characterization of the resulting copolymers was performed by Η
NMR and GPC. The results obtained are listed in Table 1.
As shown in Table 1, proteinoids having Asp and/or Glu as the second monomers and Phe and/or Tyr as the first monomers were obtained in high yield from the polymerization initiated with BzNH2 at the ratio of [M]/[I] = 5 (No. 2-1 to 2-7). The GPC curve (Figure 1, for poly (Asp.Bz-co-Phe), from which a polydispersity of 1.91 was determined. Similar molecular weight distributions were observed for other copolymers.
Polydispersity is defined herein as the molecular weight distribution of a sample. The distribution is assigned a numerical value derived from the molecular weight (MW) divided by the molecular number (Mn). The polydispersity value for a homopolymer is 1 because the molecular weight is equal to the molecular number. Any polymer with a polydispersity value of 1 is considered to have a very narrow distribution. A polymer with polydispersity value of 1.6 to 1.7 is considered to have medium distribution. A polymer with a polydispersity value of 2.0-2.1 is considered to have a broad distribution.
The homopolymerization of NCA of Asp.Bz and the copolymerizations of NCAs of Asp.Bz, Glu.Bz, Phe, and Tyr were also carried out using MeBzNH2 as initiator (No. 2-11, 2-15, and
2-16). Similar results were obtained for reactions initiated by
BzNH2.
Figure imgf000022_0001
Example 4 : Preparation of Proteinoids by the NCA Method
Using α-Methyl Tyrosine Ester as Initiator
This example illustrates the method of conducting NCA polymerizations, using ce-methyl tyrosine ester (Tyr.Me) as the initiator. The reaction conditions are essentially the same as described in Example 4 except tetrahydrofuan (THF) solvent was used. The results are listed in Table 2.
Figure imgf000023_0001
It was found that the initiation by Tyr.Me is very fast (No. 2-17 to 2-20) and all the NCA has been converted after 2 hours. From GPC data, it was observed that the molecular weight of the polymer increased with increasing ratio of [M] / [Tyr.Me] and the polydispersity is quite narrow. The e: stence of a Tyr.Me residue in the polymers was confirmed by Η NMR spectra. In conclusion, Tyr.Me is a novel and effective initiator for the polymerization of amino acid NCA's.
Sample No. 2-13 represent a polymerization initiated with β-alanine and terminated wit: succinic anhydride. As β- aaanine is insoluble in most organic solvents, the reaction was carried out in refluxing THF. As a result, the polydispersity of the polymer obtained was broader than that of the polymers initiated by Tyr.Me.
Example 5: Preparation of Proteinoids
bv the DPPA Method (#1)
This is an example of a direct polycondensation of
Asp.Bz in the presence of DPPA and triethylamine (TEA) as a base under various polymerization conditions ((a), (b), (c), and (d)).
The molecular weight of the polymers, as well as polydispersity, was evaluated in each case by GPC. The polymers were characterized by IR and NMR spectroscopy.
Asp.Bz was prepared by the esterification of L-aspartic acid as follows: L-aspartic acid (26.6 g, 0.2 mole) was suspended in 300 ml of freshly distilled benzyl alcohol in a 500 ml round bottom flask, fol±owed by addition of 45 ml of concentrated hydrochloric acid (12N). The mixture was heated up to 60ºC under vigorous stirring for 30 minutes. Thereafter, the reaction solution cooled to room temperature. Triethyl amine (about 56 ml) was added to neutralize (to a pH of about 7) the solution. The crude product was collected by filtration, washed with ethanol and acetone, dried in vacuo, and crystallized twice from hot water. 18 g of product was obtained (% yield = 44%). M.pt = 217°C.
Commercial DPPA was used without further purification. TEA was distillated before use. Solvents for polymerization were purified by conventional methods. The direct polycondensation of Asp.Bz was carried out by stirring a dimethyl formamide (DMF) solution of the monomer in the presence of DPPA and TEA. The mixture was stirred for 1 h at 0-10°C followed by stirring at room temperature for two days. The resulting polymer was precipitated in a large amount of water, collected by filtration, and then dried in vacuo. a. Effect of Monomer Concentration
Listed in Table 3 are the results for the polymerization of Asp.Bz in DMF at room temperature for two days. Poly (Asp.Bz) s were obtained from there direct polycondensations in high y ield.
The molecular weight of the polymers was found to be dependent on the concentration of the monomer [M]. Low molecular weight polymers with broad distribution were obtaine^ from a low [M] (Figure 2, curve A). On the other hand, when [M] was greater than 0.2 g/mL, a polymer with a bimodal molecular weight distribution was obtained (Figure 2, curve B). The lower molecular we it oligomers (-1000) may be due to an intramolecular termination between the terminal amino and the β-carboxylic groups. After several reprecipitations from THF/methanol, a polymer with a higher molecular weight (Mn = 22,000) and narrow polydispersity (Mw/Mn = 1.68) was successfully isolated from the polymer mixture prepared at [M] = lg/mL. The separation was also performed using GPC column with Bio-Beads.
Figure imgf000025_0001
b) The polymer was collected by filtration after polymerization for 2.5 days.
c) The values in parentheses are molar percentages. b. Effect of Reaction Time and Temperature
The yield of the resulting polymer increased with the reaction time: 75% conversion in 5 h and 95% in 4 days (Figure 3, curve A). The molecular weight of the resulting polymer also increased with time in the initial phase (up to 4 h) and then became almost constant (Figure 4). The polymerization decreased with increasing temperature (Figure 3, curve B). Polymers obtained at 60 and 80°C were of yellow color and insoluble in THF but soluble in DMF and DMSO. This may be due to the formation of an imide ring which has been reported to occur during thermal polycondensations of aspartic acid. c. Effect of Molar Ratios [DPPAl / [M] and [TEA] / [M]
The dependence of the yield and the molecular weight of the polymer on the molar ratios of [DPPA] / [M], as well as [TEA] / [M], was investigated (Table 4). The highest yield was obtained at a [DPPA] / [M] of 1.3 and a [TEA] / [M] of 2.3 (Figure 5). These observations are in agreement with the results reported by Nishi et. al. Higher molecular weight products were obtained in the range of [DPPA] / [M] = 1.3-2.0 and [TEA] / [M] = 2.0-3.0, respectively.
Figure imgf000027_0001
d. Effect of Solvent
A comparison of the polymerizations in different εolvents is shown in Table 5. It can be seen from this table that the yield and the molecular weight of the polymer are influenced by the solvents used. Higher yields were obtained in DMF while higher molecular weights were obtained in THF and in bulk. On the other hand, the polymerization in dioxane gave a lower molecular weight product, and therefore is preferred.
Figure imgf000028_0001
Example 6: Preparation of Proteinoids
by the DPPA Method (#2)
Copolymerizations of Asp.Bz with other amino acid monomers such as γ-benzyl glutamate (Glu.Bz), β-alanine (Ala), Phenylalanine (Phe), and O-benzyl tyrosine (Tyr.OBz) in the presence of DPPA were carried out using the same procedure as that for the homopolymerization of Asp.Bz (Example 5). Random copoly (amino acids) were obtained in high yield (> 77%) as shown in Table 6. This indicates that the copolymerization of amino acids using DPPA is a useful approach to copolypeptide synthesis. Bimodal molecular weight distributions were also observed in these cases similarly to the homopolymerization of Asp.Bz.
Figure imgf000029_0001
Example 7 : Reductive Debenzylation of Proteinoids
Produced by the DPPA Method
The example illustrates a preferred method for the removal of benzyl protective groups in poly (Asp.Bz) and poly (Glu.Bz) by catalytic hydrogenation.
The hydrogenation of the polymers was carried out according to the following procedure: To a solution of the polymer in THF/methanol (1:1, v/v), Pd on active carbon (10%) was added in the amount of 1/10 of the polymer weight. After the replacement of air by nitrogen, hydrogen gas was introduced into the system and maintained with a balloon. The reaction mixture was stirred at room temperature overnight. After removing the catalyst by filtration and concentrating the solution, the mixture was poured into a large amount of petroleum ether to precipitate the polymer. The polymer obtai ed was then dried in vacuo.
The completion of the hydrogenation was confirmed by Η NMR of the polymer. Ii αost cases, useful water-soluble polymers were produced. The hydrogenation is an effective and clean procedure for benzyl group removal.
Example 8: Preparation of Empty Proteinoid- carriers
with Glu, Asp, Tyr, Phe Proteinoid
This Example illustrates a method for the preparation and cleaning of empty proteinoid carriers.
PROCEDURE
1. Reagents:
a. Proteinoid powder prepared as described in Example 1 b. Anhydrous citric acid (USP)
c. Gum acacia NF
d. Deionized water
e. Glatial acetic acid
2. Equipment:
a. Ph meter
b. Water bath, 40°C
3. Preparation of Solutions:
a. Proteinoid solution - Dissolve 100mg proteinoid in 1ml deionized water (or multiples thereof). Filter through a Whatman #1 filter paper (if necessary) and keep at 40°C in a water bath. This is solution A. b. 1.7 N citric acid with 0.5% acacia - Dissolve 5g of acacia and 109g of citric acid in 1 liter deionized water. Incubate at 40°C. This is solution B.
Preparation of Proteinoid carriers:
a. Add all of solution A to solution B rapidly in one step while swirling solution B by hand, in a 40°C water bath. Example 9: Preparation of Murine IgG Monoclonal Antibody- containing Proteinoid Carrier
This experiment describes encapsulation of anti-reovirus monoclonal antibody (mAb) 9BG5, an mAb directed against the sigma-1 gene product (Hemaglutinin, HA3) of the Reovirus Type
3. HA3 binds to the cell surface receptor for Reovirus type 3, and mAb 9GB5 interferes with viral binding to the receptor.
Mouse IgG monoclonal antibody 9BG5 was prepared and purified as described W.V. Williams et al. (1991) J. Biol. Chem., Vol. 266(8), pages 5182-5190, as well as references cited therein, using a purified Reovirus type 3 preparation (W.V. Williams et al. (1988) Proc. Natl. Acad. Sci. U.S.A, Vol. 85, pages 6488-6492). The purified 9BG5 used in this Example had a protein concentration of 1.5 mg/ml in phosphate buffered saline (pH 7.2).
Proteinoid carriers encapsulating mAb 9BG5 were prepared having final concentrations of Glu/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Glu, Asp, Tyr, and Phe in the reaction mixture) 50 mg/ml, mAb 0.7 mg/ml and gum arable 0.5% in 0.85 N citric acid. Empty proteinoid carriers were prepared to contain the same final concentrations, except mAb was omitted. Aliquots (0.5 ml), in duplicate, of both mAb and empty proteinoid carriers preparations were centrifuged at 5000 RPM. Pellets and supernatants were frozen prior to analysis by Western blotting to determine antibody encapsulation efficiency.
Figure 6 is an x-ray film of a western blot analysis of purified mAb 9BG5, empty proteinoid carriers (no mAb added), and proteinoid carriers containing 9BG5. The analysis was done by immunoblotting with anti-mouse IgG which specifically reacted with mAb 9BG5. The lanes correspond to the following:
Lane Sample
1 2 μg 9BG5 mAb
2 1 μg 9BG5
3 0.25 μg 9BG5
MW molecular weight markers
4 Empty proteinoid carrier supernatant after encapsulation
5 Empty proteinoid carrier pellet 6 mAb containing supernatant after encapsulation
7 mAb containing protein carrier pellet
The data indicates that the 9BG5 proteinoid carriers contained about 40% of the mAb in the pellet and the remaining 60% did not incorporate in the proteinoid carriers and was left in the supernatant. The empty proteinoid carriers did not contain antibody in the supernatant or the pellet as was expected.
The relative mobility (molecular weight) of the pure mAb is slightly different than the mAb in the proteinoid carriers. This is most likely due to different salt concentrations in the samples, because the encapsulation process employed 0.8 M salt solution.
Example 10: Effect of Additives on Stability of Proteinoid
Carriers with Encapsulated Murine mAb 9BG5
Various proteinoid carrier formulations were screened, with or without additives, to determine optimal carrier-forming conditions and concentrations of mAb required for carrer formation.
The mAb 9BG5 preparations used to prepare the encapsulated proteinoid carriers had a protein concentration of approximately 2 mg/ml in phosphate buffered saline.
Final proteinoid concentration was 50 mg/ml and 5% (w/w) gum acacia ("gum") or gelatin ("gel"). All proteinoid carriers were prepared in 0.85 N citric acid. Empty carriers were included for use as controls, and they were prepared in the same manner with the omission of mAb. Duplicate (0.5 ml) aliquots of proteinoid carrier suspension were centrifuged at 5000 RPM. Pellets and supernatants were frozen in dry ice prior to analysis.
Table 7 lists samples that were prepared. Numbers in parenthesis indicate amount of mAb added.
Figure imgf000033_0001
In order to test resistance to freeze and thawing on the integrity of the proteinoid carriers containing mAb, one of each pair of duplicate pellets were washed by gentle resuspension in 0.25 ml of 0.85 N citric acid. The pellets were then analyzed next to the unwashed pellets to test whether any mAb was lost in the washing.
The samples were analyzed by conventional Western blotting as described in Example 9. Pellets were dissolved in sodium dodecyl sulfate with 0.05 N NaOH and analyzed under reducing conditions (breaks up the mAb into 50 kDa and 25 kDa bands). Aliquots (50μl) of supernatants were analyzed under non- reducing conditions (expected intact 50 kDa mAb). This was done to determine differentially whether the mAb left behind is denatured or intact.
As can be seen from the X-ray film from the Western Blots (Figure 7), pellets of samples 9 and 10, and 11 and 12 contain between 5 and 10 μg of mAb. The washed samples did not lose any significant amount of mAb, suggesting that the proteinoid carriers remained intact after freeze-thawing.
The supernatants of samples 9 and 11 had no significant amount of mAb, indicating that unincorporated material was lost during preparation.
Sample 17 had some mAb encapsulated which was lost after washing (see number 18). This sphere preparation was not resistant to freeze-thawing. Additionally, a band at a MW of 150 kDa for sample 17 supernatants indicates that a significant amount of mAb is left behind after proteinoid carrier formation.
Based on these results, it appears that the mAb remains intact and therefore the encapsulating procedure does not degrade it. The empty proteinoid carrier controls did not produce any bands, as expected because they have no mAb.
Example 11: Efficacy of Encapsulated Murine
IgG Monoclonal Antibody
In this Experiment, a mAb 9BG5 proteinoid carrier preparation and unencapsulated mAb 9BG5 were evaluated in rats. The mAb 9BG5 (1 mg/ml), prepared as described in Example 9, was encapsulated in Glu/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Glu, Asp, Tyr, and Phe in the reaction mixture) protein carrier formulation with gum arabic. The mAb proteinoid carriers suspension contained 0.25 mg/ml mAb and 50 mg/ml proteinoid in 0.85 N citric acid-0.5% gum. Empty proteinoid carriers were prepared similarly, but did not contain mAb. Since 30% of the mAb was found to be encapsulated, the mAb proteinoid carriers were estimated to contain 0.075 mg/ml mAb and this value was used to determine dosages. The mAb proteinoid carriers were examined microscopically and appear to be a fairly homogeneous preparation.
For animal dosing, appropriate aliquots of proteinoid carriers were centrifuged at 5000 RPM for 15 minutes, and pellets were resuspended in 1.0 ml of 0.85 N citric acid-0.5% gum.
A purified mAb solution (0.95 mg/ml mAb in 0.85 N citric acid-0.5% gum) was used for oral gavage. This solution was prewarmed to 40ºC prior to administration. For IV administration, a purified mAb solution (1 mg/ml mAb in phosphate buffer saline) was used.
The amounts and administration routes employed in the experiment are as follows:
1. Empty proteinoid carriers (no mAb) : 1 ml aliquot containing
50 mg empty proteinoid carriers by oral gavage (rats # 2312 and 2313).
2. mAb 9BG5 proteinoid carriers: 3.7 mg mAb/ kg body weight of rat by oral gavage (rat # 2287, 2288, 2290, and 2291).
3. unencapsulated mAb 9GB5 : 0.73 mg/ kg body weight of rat by intravenous administration (rats #2292, 2293, and 2311).
4. unencapsulated mAb 9BG5 : 3.7 mg/ kg body weight of rat by oral gavage (rats #2314 and 2315).
Baseline blood samples (1 ml aliquots) were withdrawn from each rat just prior to dosing ("0" time). After dosing, blood samples were drawn at 1 h, 6 h and 24 h. The blood samples were processed immediately and sera were stored frozen at -20°C.
Thawed serum taken from the experimental animals were analyzed by conventional ELISA techniques, in triplicate, using purified reovirus type 3 and VLSH dimeric peptides immobilized in multi-well plates (W.V. Williams et al (1991) J. Biol. Chem., Vol. 266(8), pages 5182-5190). Control plates included wells having no immobilized reovirus and VLSH peptides to which mAb (1mg/ml) was added. VLSH peptide (W.V. Williams et al. ibid, Table 1) is a synthetic variant of VL peptide, the latter which corresponds to a portion of the light chain variable CDR II re on of 87.92.6 antibody. The 87.92.6 antibody displays idiotypic and anti-idiotypic behavior towards reovirus type 3 receptor and mAb 9BG5, respectively (W.V. Williams et al. ibid). The bound protein content of each well were measured by standard protein methods, e.g., Lowry method, and the results for each multi-well plate are shown in Figures 8(a-c), respectively.
Figures 8 (a-c) illustrate the levels of serum proteins which bound to immobilized reovirus type 3 and VLSH as detected by measurement protein concentration. These Figures show that the serum levels of bound proteins, after 24 hours post-dosing, were highest for animals orally administered mAb proteinoid carriers and animals administered unencapsulated mAb by the IV route. Lower levels of bound serum proteins were found in animals orally adminstered uncapsulated mAb. Serum taken from the animals receiving empty proteinoid carriers (no mAb) showed non-specific serum IgG protein binding, as expected, under the assay conditions.
Figure 9 show mAb binding under conventional ELISA procedures using immobilized reovirus type 3 and VLSH proteins.
Serial dilutions of mAb treated with 0.85 N citrate-0.5% gum
(Figure 9(a) or phosphate buffered saline (Figure 9 (b) were employed. The Figures show that the bound protein levels were higher for mAb in citrate buffer than for mAb in phosphate. Without being bound by any theory of operation for this invention, it is believed that the binding enhancement may be due to changes in the three dimensional conformation resulting from citrate-protein binding.
In summary, serum levels of mAb, as reflected by the absorbance of bound proteins, were greater in animals receiving encapsulated mAb by the oral route or unencapsulated mAb by the IV route, than an animal receiving orally administered unencapsulated mAb.
Example 12 : Preparation of Proteinoid carrier containing
Heparin
This Example describes a method for the preparation and cleaning of heparin proteinoid carriers.
PROCEDURE
1. Reagents:
a. Proteinoid powder prepared as described in Example 1 b. Heparin
c. Anhydrous citric acid (USP)
d. Gum acacia NF
e. Deionized water
f. Desiccant
g. Liquid nitrogen 2. Equipment:
a. Magnetic stirrer
b. Buret
c Microscope
d. Clinical centrifuge
e. Dialysis membrane tubing (Spectrum 6, 10 mm, 50,000 M.W. Cutoff)
f. pH meter
g. Lyophilizer (Labconco #75035)
h. Lyophilizing flasks (150-300 mL)
i. Rotating shell freezer
j. Isopropanol/dry ice bath or liquid N2
k. Mortar and pestle
l. Storage containers (500 mL)
m. Eppendorf pipet (0-100 uL)
n. Plastic closures for dialysis tubing (Spectrum) o. 2 mL syringe with 0.45 um Acrodisk
3. Preparation of Solutions:
a. Proteinoid Solution A* (80 mg/ml) :
Dissolve 160 mg proteinoid in 1 ml of deionized water. Using a 2 ml syringe fitted with a 0.45 um Acrodisk, the proteinoid solution was filtered into a 10 ml test tube and kept at 40° C. b. Solution B (1.7 N citric acid with 1% gum):
Dissolve 10 g of gum acacia and 109 g of citric acid in 1 liter of deionized water.
Solution C (Heparin solution):
Dissolve heparin in Solution B at 150 mg/mL and keep at
40° C.
* or multiples thereof 4. Preparation of Proteinoid carriers:
a. Add all of solution A to solution C quickly while swirling solution C slowly, by hand, in a 40°C water bath.
5. Dialysis of Heparin Proteinoid carriers:
It has been found the presence of citric acid in the encapsulated proteinoid carriers interferes with a subsequent lyophilization process. Hence, proteinoid carrier encapsulates prepared with citric acid solutions are preferably dialyzed against 5% acetic acid solution for at least two hours with at least four changes of the dialysis solution to remove citric acid by an exchange process. Thus,
a. Transfer the suspension with a syringe (no needle) to dialysis tubing and seal with plastic closures. Tubing should be no more than 70% full. b. Discard any amorphous material sedimented and/or aggregated on the surface. c. Dialyze the proteinoid carrier suspension against acetic acid solution (using 20 mL of acetic acid solution per ml of proteinoid carrier suspension) while stirring the acetic acid solution with a magnetic stirrer. d. Replace the acetic acid solution every hour. Continue dialyzing for a total of 3 hours. 6. Lyophilization:
a. Add one part of 50% trehalose (Sigma Chemical Co., St.
Louis, MO, USA) into nine parts of dialyzed proteinoid carrier solution. Flash freeze proteinoid carriers in a freeze-drying flask using the shell freezer adjusted to rotate at ca. 190 rpm and immersed in a liquid nitrogen bath. b. Freeze dry for 24 hours or until dry as evidenced by lack of self-cooling. c. Record weight of dry proteinoid carriers. d. Grird to a fine powder with mortar and pestle. e. Transfer proteinoid into an amber container, seal with desiccant, and store at room temperature.
7. Resuspension:
a. Weigh the lyophilized powder and calculate the amount of proteinoid in the powder. b. Add aqueous 0.85 N citric acid into the lyophilized powder at 40°C. The final concentration of proteinoid in solution is 80 mg/ml.
Example 13: Preparation of Insulin-containing Proteinoid
Carrier
This Example illustrates a method for the preparation of insulin proteinoid carriers.
PROCEDURE
1. Reagents :
a. Proteinoid powder
b. Anhydrous citric acid (USP)
c. Gelatin (USP)
d. Porcine insulin (Novo Nordisk)
e. Deionized water (USP) 2. Equipment:
a. Water bath b. 0.2 micron Acrodisk filter
c. Sterile syringe (10cc)
d. Glass or plastic vessel of appropriate volume for desired amount of proteinoid carrier-solution.
3. Preparation of Solutions:
a. 1.7 N citric acid with 5.0% gelatin:
Dissolve 109 mg anhydrous citric acid and 50 mg gelatin per 1 ml of deionized water at desired volume** and incubate in water bath at 40°C until gelatin is completely dissolved. This may be prepared and stored at 40°C for later use. b. Insulin solution:
Dissolve 12 mg insulin per 1 ml of 1.7 N citric acid with 5% gelatin at 40°C at desired volume. c. Proteinoid solution:
Dissolve 100 mg proteinoid per 1 ml deionized water at room temperature and desired volume. Using syringe and
0.2 micron Acrodisk, filter the solution to ensure a clear liquid and incubate in a water bath at 40°C. See Section 5b. 4. Preparation of Proteinoid carriers:
a. Proteinoid solution and insulin solution are combined at equal volumes sufficient to produce the final desired volume of proteinoid carriers. b. Rapidly add the filtered proteinoid solution to the insulin solution at 40°C while simultaneously and constantly swirling the insulin solution to ensure a ** Proteinoid and Insulin solutions should each be prepared at one-half the total volume of the final microsphere solution desired. thorough mixing.
Example 14: Procedure for Preparation of Erythropoietin
Containing Proteinoid carriers
Encapsulation of human erythropoietin (EPO) in proteinoid carriers was performed in the same manner described in
Example 13. EPO was obtained from Genetic Institute (Cambridge,
MA, USA, now available from Amgen Corp., Thousand Oaks, CA, USA).
A solution of Gln/Asp/Tyr/Phe (1: 1:1 mole ratio of Gln, Asp, Tyr, and Phe in the proteinoid reaction mixture) proteinoid and a 150 ug/mL EPO solution in 1.7 N citric acid with 1% gum was used in preparing the EPO-containing proteinoid carrier.
Example 15: Evaluation of Erythropoietin-containing
Proteinoid Carrier
In this Example, an EPO-containing protein carrier, prepared as described in Example 14, was evaluated in rats. An
EPO experimental synopsis is given below.
Rats weighing 50-200 grams are anesthetized with ketamine (8.5mg/kg) and thorazine 3.75mg/kg) with intramuscular injection. The rat is then administered either unencapsulated erythropoietin or encapsulated erythropoietin by oral gavage. In brief, an 8 french nelaton catheter is inserted down the esophagus of the rat until the 10cm mark on the catheter is even with the incisors. The test or control solution is drawn up into a syringe and attached to the catheter. Holding the animal upright, the solution is expressed into the stomach of the rat. The experimental results are summarized in Figures 10-12.
ERYTHROPOIETIN EXPERIMENTAL SYNOPSIS
Batch Dose Rats Responding Comments
Control 15μg/kg 0/4 Fasted 15 hours.
251<3K 15μg/kg 0/4 Access to bedding.
254<3K 15μg/kg 2/4 Gavaged
Control 15μg/kg 0/2
251<3K 15μg/kg 0/2 Fasted 36 hours.
254<3K 15μg/kg 1/4 5% sucrose.
270K 15μg/kg 1/3 No bedding.
270G 15μg/kg 3/3 Gavaged.
Control 15μg/kg 1/5 Fasted 24 hours.
264CP 15μg/kg 1/4 Access to bedding.
270G 15μg/kg 1/6 Gavaged.
Control 10μg/kg 0/5 Fasted 24 hours. 270G 10μg/kg 3*/6 No bedding.
Control 30μg/kg 0/3 Fasted 24 hours. Control 60μg/kg 1/4 No bedding.
270G 30μg/kg 1/3 Direct injection 270G 60μg/kg 1/4 into the stomach.
Control 50μg/kg 0/3
Control+
Pepsin 50μg/kg 0/4 Direct injection 270G 50μg/kg 2/4 into the intestine. 270G +
Pepsin 50μg/kg 0/4
Control 100μg/kg 1/5 Multiple Dosing 270G 100μg/kg 1/5 (5 dosing intervals I.V. 50μg/kg 2/2 at t 1/2)
S.C. 50μg/kg 2/2 Gavage by stomach tube.
*Rats were foaming at nostrils.
Figure 10 illustrates levels of erythropoietin (EPO) detected in rat serum taken from rats administered Gln/Asp/Tyr/Phe proteinoid carrier encapsulated EPO (15μg EPO/kg body weight) and encapsulated EPO (15μg EPO/kg body weight) at t = 0.5, 1, and 2 hours. Serum erythropoietin levels were determined over time with an erythropoietin enzyme immunoassay kit (Amgen, Thousand Oaks, CA, USA). The results show that EPO serum levels in rats administered erythropoietin proteinoid carriers were relatively higher at all time points compared to rats (control) which received unencapsulated material. At t = 2 hours, the EPO levels remained at approximately 300 pg/mL serum in rats administered erythropoietin proteinoid carriers while the control rats had undetectable EPO levels.
Figure 11 illustrates EPO serum levels in rats that were administered either erythropoietin (50μg/kg) or Gln/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Gln, Asp, Tyr, and Phe in the reaction mixture) proteinoid carrier encapsulated erythropoietin (50μg/kg) directly into the proximal duodenum. Serum erythropoietin levels were determined over time with the aforementioned erythropoietin enzyme immunoassay kit. The results show that EPO serum levels in rats administered erythropoietin proteinoid carriers steadily increased at a rate of approximately 50 pg/mL per hour over a range of two hours. In contrast, rats (control) which received unencapsulated EPO had EPC levels peaked at 100 pg/mL at 1 hour following administration and steadily decreased to about 50 pg/mL at the end of 2 hours.
Figure 12 illustrates EPO strum levels in rats who were orally gavaged with either Gln/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Gln, Asp, Tyr, and Phe in the reaction mixture) p-roteinoid carrier encapsulated or unencapsulated erythropoietin (100μg/kg); or received a subcutaneous injection of either 2μg/kg or 10μg/kg. Serum erythropoietin levels were determined over time with the aforementioned erythropoietin enzyme immunoassay kit. The results show that EPO serum levels in rats (#640-645) orally administered erythropoietin proteinoid carriers were relatively higher up to t = 2 hours, compared to rats (EPO) which received unencapsulated material.
The results obtained in this Example provide evidence that proteinoid encapsulation markedly improved the oral bioavailability of EPO. Example 16: Preparation of Calcitonin-containing
Proteinoid carrier
Encapsulation of salmon calcitonin in proteinoid proteinoid carriers was performed in the same manner described in Example 13. Calcitonin, a peptide hormone which acts predominantly on bone to lower serum calcium concentration, was obtained from Sandoz (Basil, Switzerland). Calcitonin proteinoid carriers were prepared by mixing a 1:1 volume ratio of a 100mg/ml aqueous solution of Gln/Asp/Tyr/Phe proteinoid (1:1:1:1 mole ratio of Gln, Asp, Tyr, and Phe used in the proteinoid reaction mixture) and a 150 ug/mL calcitonin solution in 1.7 N citric acid solution with 1% gum acacia, as described in Example 13. The efficiency of calcitonin encapsulation was approximately 40%. Calcitonin concentration was determined directly by HPLC after dissolving the calcitonin proteinoid carriers in 60% aqueous acetonitrile.
Example 17: Evaluation of Calcitonin-containing
Proteinoid carriers in Monkeys
In this Example, the calcitonin proteinoid carriers, prepared as described in Example 16, were evaluated in cynomolgus monkeys. Male cynomolgus monkeys weighing 4-5 kg were fasted overnight, anesthetized (approximately 10mg/kg ketamine HCl) and placed into a primate restraint chair for dosing and blood sampling. A single oral dose of calcitonin proteinoid carriers (0.25 mg/kg body weight) was administered to each of four monkeys by nasogastric gavage. The dosage was based on the body weight taken on the morning of dosing. Blood samples were collected from saphenous vein catheters at hourly intervals, starting at t = 0 prior to administration of the proteinoid carriers, and hourly, from 1 to 7 hours post-dose for serum calcium determination. The hypocalcemic response following oral calcitonin administration was used as an index of pharmacological response. Serum calcium concentrations were quantitated by a conventional O-cresolphthalein complexone method.
Figure 13 demonstrates the response obtained in cynomolgus monkeys following naso-gastric gavage of microencap sulated calcitonin. Significant changes from baseline serum calcium concentration were observed. Six hours following dosing, serum calcium concentrations decreased by 13 μg/ml. A significant pharmacological response was still apparent seven hours after the administration of calcitonin proteinoid carriers.
Example 18: Evaluation of Calcitonin-containing Proteinoid
Carriers in Rats
In this Example, the calcitonin proteinoid carriers prepared in accordance with Example 16 are evaluated in fasted male Spraque Dawley rats weighing 100-150g. Calcitonin proteinoid carriers and calcitonin were administered by either oral gavage or intraduodenal injection. The rats are divided into the following groups:
1. calcitonin proteinoid carriers: 60 ug calcitonin/kg body weight by oral gavage (3 rats);
2 . calcitonin proteinoid carriers: 3 ug calcitonin/kg body weight by intraduodenal gavage (3 rats);
3. calcitonin: 60 ug calcitonin/kg body weight by oral gavage (3 rats) (Control).
calcitonin: 3 ug calcitonin/kg body weight by intraduodenal gavage (3 rats) (Control).
Oral gavage dosing of rats is performed. Calcitonin proteinoid carriers are prepared immediately prior to dosing and Groups 1 and 2 each receive an appropriate dosage of the proteinoid carrier suspension. Groups 3 and 4 receive the unencapsulated calcitonin (no proteinoid carriers). Approximately 0.5 ml of blood is serially withdrawn from the tail artery of each rat just prior to dosing ("0" time) and 1 h, 2 h and 3 h post-dosing Serum from the blood samples are stored at -20°C for serum calcium concentration determination.
Figure 14 is the serum concentration-time curve for orally administered microencapsulated calcitonin and unencapsulated calcitonin in rats. Experimental results in rats demonstrate a significant increase in pharmacological response (i.e., decreasing serum calcium levels) when proteinoid encapsulated calcitonin is compared to the unencapsulated vehicle control group. One hour after dosing, serum calcium concentrations decreased 23 μg/ml in the rats receiving encapsulated calcitonin compared to a decrease of only 6.5 μg/ml in the control group. Furthermore, the responses were dose-dependent (data not shown).
The results of intraduodenal injection of encapsulated or unencapsulated calcitonin in rats is shown in Figure 15. The results demonstrate a time-dependent decrease in serum calcium levels for the encapsulated preparation. The control group showed no response. One hour after intraduodenal administration, serum calcium levels in the calcitonin proteinoid carrier group decreased by 18μg/ml, whereas unencapsulated calcitonin was unchanged. These results indicate that transmembrane transport of calcitonin is enhanced by proteinoid encapsulation.
The results obtained in this Example and in Example 17 provide evidence that proteinoid encapsulation markedly improves the oral bioavailability of calcitonin. The data also indicate that the oral drug delivery system is not species-dependent.
Example 19: Preparation and Evaluation of Factor IX-containing Proteinoid Carrier
Factor IX is a vitamin K-dependent blood coagulation proenzyme, MW 56 kD. Factor IX deficiency, known as hemophilia B, occurs in approximately 1 out of every 25,000 males. To date, treatment of this disorder is accomplished by intravenous administration of Factor IX, although a recent report details efforts to supplement by subcutaneous injection (Thompson (1986) Blood, Vol. 67(3), pages 565-572).
Encapsulation of Factor IX (FIX) in proteinoid carriers was performed, following the procedure described in Example 13, by mixing (1:1 v/v) 100 mg/mL of Glu/Asp/Tyr/Phe proteinoid
(1:1:1:1 mole ratio of Glu, Asp, Tyr, and Phe used in the proteinoid reaction mixture) in deionized water and an aqueous solution of FIX. Two proteinoid carrier suspensions were prepared and evaluated in vivo separately as described in Examples 20 and 21.
FIX proteinoid carrie αspension A contained 50 mg/ml of proteinoid and 500 U/ml FIX (FIX is available from the
American Red Cross, Rockville, Maryland, USA) sσϊution containing 4% acetic acid, 2% gum acacia, 0.2% PEG 14 (available from Union
Carbide, Danbury, CN, USA), 14 mM CaCl2, final pH 3.81.
The second suspension, FIX proteinoid carrier suspension B, contained 50 mg/mi proteinoid and 116 U/ml FIX solution containing 3.8% acetic acid, 1.5% gum acacia, 0.15% PEG 14, 11 mM CaCl2, final pH 4.58.
The stability of FIX proteinoid carrier preparations was assessed over a sho: time course in vitro. The protein carriers encapsulating FIX were examined by optical microscopy and laser light scattering. Aliquots of proteinoid carrier suspension were withdrawn every 30 minutes for 1.5 hours, FIX proteinoid carriers were isolated by centrifugation at 4500Xg and dissolved in activated partial thromboplastin time (APTT) assay buffer (0.05M histidine-0.01M NaCl-0.1% bovine serum albumin0.01% TWEEN-40, pH 7.47) to release soluble FIX and proteinoid. Quantitation of FIX activity by APTT employed both FIX standards (0.025, 0.05, and 0.1 U/ml) and "empty" proteinoid carrier suspension as control. APTT assay kits are commercially available, e.g. Sigma Diagnostics (St. Louis, MO, USA).
Based on the above analysis, it was determined that FIX proteinoid carriers of greater stability are obtained by encapsulating FIX at a higher pH, e.g., pH 4.9. Furthermore, the efficiency of encapsulation is approximately 20% of available FIX units and activity levels remain constant for at least 1.5 hours when FIX proteinoid carrier pellets are stored at about 4ºC.
Example 20: Evaluation of FIX-containing Proteinoid carriers
(A) in Rats
In this Example, FIX proteinoid carrier suspension A, prepared as described in Example 19, were evaluated in male Sprague Dawley rats (ave. weight 300g). Appropriate aliquots of suspension were centrifuged at 4500Xg to pellet the FIX protein carriers, which were subsequently resuspended in the same buffer for animal dosing. The rats are divided into two groups as follows:
1. Oral FIX proteinoid carriers (FIX sph PO) : 2709 U FIX/kg body weight by intragastric gavage (4 rats);
2. Intravenous FIX (no proteinoid carriers) (FIX IV): 200 U/kg body weight by intravenous injection. 32 rats received 0.7 ml FIX in 0.11 NaCl-0.02M sodium citrate, pH 6.85 by tail vein injection.
The FIX proteinoid carrier suspension and solution are prepared immediately prior to dosing. One ml of blood was withdrawn from each rat just prior to dosing ("0" time) and 1 h, 2 h and 4 h (post-dosing), a citrate anticoagulant was added to the blood, and plasma from the blood samples were stored at -70°C.
Plasma samples were assayed by a modified APTT assay using FIX coagulated deficient plasma (assay kit is available from Ortho Diagnosis (Raritan, New Jersey, USA). Changes in clotting times were calculated by subtracting individual baseline (0 hr) values from subsequent clotting time values. The data shown in Figure 16 are the mean values for a given group. Values below baseline indicate the presence of exogenous FIX.
As shown in Figure 16, significant amounts of FIX was delivered to blood via oral administration of FIX proteinoid carriers. The relative plasma level is lower in the FIX proteinoid carriers group, however the dimunition in clotting time at 0.5, 1.0 and 2.0 hours is notable. This is achieved by oral dosing with approximately 14 times the IV dose. Moreover, these results are particularly interesting since Factor IX is an acid labile protein whose half-life is approximately less than one hour at 37ºC at pH 5.0. The FIX proteinoid carriers in this experiment were at pH 3.81 and encapsulated 14.8% of the available FIX units during preparation. The results support that FIX proteinoid carriers remain viable in the GI tract to facilitate delivery. Example 21: Evaluation of FIX-containing Proteinoid Carriers
(B) in Rats
In this Example, FIX proteinoid carrier suspension B, prepared as described in Example 19, were evali ed in male Sprague Dawley rats (ave„ weight 300g). Resuspended FIX proteinoid carriers were prepared as described in Example 20.
The rats are divi led into two groups as follows:
1. Oral FIX proteinoid carriers (FIX sph PO) : 1006U FIX/kg body weight by intragastric gavage (5 rats).
2. Intravenous FIX (no proteinoid carriers) (FIX IV) : 185 U/kg body weight by intravenous injection. 3 rats received 0.3 ml FIX in 0.11 NaCl-0.02M sodium citrate, pH 6.85 by tail vein injection.
3. Oral FIX (no proteinoid carriers) (FIX unencap PO) : 2760U FIX/kg body weight by intragastric gavage. 4 rats received
1.0 ml of FIX in saline solution containing 3.8% acetic acid, pH 6.85.
The FIX proteinoid carrier suspension and solutions were prepared immediately prior to dosing. Plasma samples were obtained and assayed as described in Example 20. Changes in clotting times were calculated by subtracting individual baseline
(0 hr) values from subsequent clotting time values. The data shown in Figure 17 are the mea~ values for a given group. Values below baseline indicate the presence of exogenous FIX. The FIX proteinoid carriers, prepared at pH 4.58, encapsulated 23.1% of the FIX units.
As shown in Figure 17, at oral dose levels of only 5 times that of the IV dose, significant oral delivery was observed. In addition, native FIX (pH 6.85) dosed at 15 times the IV dose level resulted in no detectable levels of exogenous
FIX in the plasma.
Thus, the results shown in this Example and in Example
20 support that oral delivery of FIX can be accomplished via the use of FIX proteinoid carriers. These proteinoid carriers appear to adequately protect FIX during transit through the GI tract and deliver FIX to the blood stream. Example 22: Preparation of alpha-Interferon (IFN)-containing
Proteinoid carrier
In this Example, a study was undertaken to evaluate the protective capability of proteinoid carriers on enzymatic degradation under simulated gastrointestinal conditions. The in vi tro stability of IFN in proteinoid carriers was examined in simulated gastric fluid (SGF) containing pepsin in 0.08 N HCl and simulated intestinal fluid (SIF) containing pancreatin in phosphate buffer. The reagents and stability assay procedure are described in the "United States Pharmacopoeia" (Vol. XXII, 1990, pages 1788 and 1789).
Preparation of IFN-containing proteinoid carriers
Encapsulation of IFN in proteinoid carriers was performed in the same manner described in Example 13. Alpha- IFN is available from a number of commercial sources. One commercial IFN product includes Roferon-A (Hoffman LaRoche). IFN proteinoid carriers were prepared with an aqueous solution of Glu/Asp/Tyr/-Phe proteinoid (1:1:1:1 mole ratio of Glu, Asp, Tyr and Phe used in the proteinoid reaction mixture), and an IFN solution containing 1.7 N citric acid solution with 5% gelatin. The IFN proteinoid carrier suspension contained 80 mg/ml proteinoid, 600 ug/ml IFN, 0.6N citric acid, and 2.5% gelatin, pH 3.0. Stability of IFN proteinoid carriers in SGF
SGF (2 ml) was added into 1 ml of IFN proteinoid proteinoid carrier suspension. The solution was incubated at 40 "C with shaking, and aliquots were taken serially after SGF addition as described in the "U.S. Pharmacocopia" (ibid). An equal volume of stopper solution (pepstatin A in phosphate buffer, was added to each aliquot immediately after sampling to stop the enzymatac degradation and to open the proteinoid carriers. The IFN concentration in all samples was then determined by HPLC. As a comparison, the stability of IFN alone in SGF was evaluated. The experiment were performed as described above, without the proteinoid carriers. As another control, the stability of IFN proteinoid carriers was evaluated in 0.08 N HCl.
Stability of IFN-containing proteinoid carriers in SIF
SIF (2 ml) was added into 1 ml IFN proteinoid carriers. The solution was incubated at 40ºCC with shaking and samples were taken serially as described in the "United States Pharmacocopia" (ibid). An equal volume of stopper solution (aprotinin and trypsin/chymotrypsin inhibitor in phosphate buffer) was added to each aliquot immediately after sampling to stop the enzymatic degradation. The IFN concentration was analyzed by HPLC.
To study the study the stability of IFN alone in SIF, 600 ug of IFN was dissolved in 0.85 N citric acid or 0.01 M phosphate buffer. SIF (2 ml) was added to 1 ml IFN solution. The solution was sampled and analyzed as described above.
Results and Discussion
(a) Protective Effects of Proteinoid carriers in SGF
As shown in Figure 18, after 1 hour of SGF incubation, approximately 50% of IFN remained intact. After incubation in SGF for 6 hours, approximately 20% of IFN was not degraded. As expected, IFN alone (in the absence of proteinoid carriers), was found to be completely destroyed by pepsin in SGF within 20 minutes.
Another control was performed using IFN alone in 0.08 N HCl. IFN alone was stable in SGF without pepsin (0.08 HCl). There was only a slight decrease after 2 hour incubation. This suggests that IFN was rather stable in HCl at pH 1.2 up to six hours (Figure 19).
The results suggest that proteinoid carriers can retard IFN from pepsin digestion, while IFN alone cannot survive in tne stomach for more than 20 minutes. These observations demonstrate the protective ability of proteinoid carriers on enzymatic digestion of protein drugs in the stomach. (b) Protective Effects of Proteinoid carriers in SIF
As shown in Figure 20, IFN proteinoid carriers were much more stable than IFN alone (in the absence of proteinoid) m SIF. IFN alone at pH 7.4 was completely degraded within 10 minutes when incubated with SIF. However, approximately 70% of the IFN/proteinoid carriers survived after 6 hours in SIF, indicating that considerable stability is provided by the proteinoid proteinoid carrier.
IFN alone was slightly more stable in SIF at pH 3 than at pH 7.4. After 6 hr incubation in SIF at pH 3, there was approximately 10% of the IFN remaining. The stability of IFN in SIF at pH 3 is attributed to the low pH, which appears to suppress enzymatic activity of the intestinal proteases. Example 23 : Evaluation of Heparin-containing Proteinoid carriers in Rats
In this Example, a study was undertaken to ascertain whether proteinoid carriers are required for protective capability or whether (1) proteinoids (soluble proteinoids- -not in carrier form) may be used and whether (2) alternative methods of carrier loading, such as incubating the therapeutic compound with preformed proteinoid carriers, are useful.
Preparation of Heparin-containing proteinoid carriers
Encapsulation of heparin in proteinoid carriers was performed in the same manner described in Example 12. Heparin
(USP grade) was used and this material is available from a variety of commercial sources including Eli Lilly (Indianapolis,
USA). Heparin proteinoid carriers were prepared, following the procedure of Example 12, using a 1:1 volume ratio of 150 mg/ml of Glu/Asp/Tyr/Phe/Orn0.5 (1:1:1:1:0.5 mole ratio of Glu, Asp, Tyr, Phe, and Orn used in the proteinoid reaction mixture) proteinoid in deionized water, and an 20mg/mL aqueous heparin solution containing 1.7 N citric acid solution and 0.5% gum acacia. The heparin proteinoid carrier suspension was dialyzed in acetic acid solution as described in Example 12. Heparin proteinoid carriers were then centrifuged at 4800Xg (15 minutes) and total heparin was measured by assaying the pellet and the supernatant with a modification of the Azure A method (Gundry et al. Amer. J. of Surgery (1984) Vol. 148, pages 191-194). Proteinoid was assayed by dissolving the proteinoid carriers with 0.1 N NaOH and measuring absorbance at 294 nm.
Preparation of heparin-spiked empty proteinoid carriers
Empty proteinoid carriers were prepared following the same procedure described above for the heparin proteinoid carriers, with the modification being that no heparin was present. The lyophilized empty proteinoid carriers were resuspended in 0.85N citric acid and 0.5% gum containing heparin at a concentration of 20 mg/ml. The amount of heparin coisolated with the proteinoid carriers was measured as described above.
Experimental Procedure
Male Spaque Dawley rats weighing approximately 350g were dosed by oral gavage or intraduodenal (ID) injection (just anterior to the pyloric sphincter and into the duodenum). Rats were dosed orally or ID with one of the following: lyophilized heparin proteinoid carriers, heparin-spiked empty proteinoid carriers, proteinoid/heparin in water, heparin in 0.85N citric acid and 0.5% gum and heparin alone in water. In both oral and ID injection experiments, weight ratios of neparin:proteinoid were constant. The total heparin dose in the oral studies was 100 mg/kg body weight; in ID injections studies, it was 50 mg/kg. The proteinoid dose was 40 mg/kg for oral gavages and 20 mg/kg for ID injections. The dosing volume was approximately 0.3 to 0.5 ml. Approximately 0.5 ml of blood is serially withdrawn from the tail artery of each rat just prior to dosing ("0" time) and 1 h, 2 h and 4 h post-dosing. Serum from the blood samples are stored at -20°C for heparin activity determination. Results and Discussion
The results obtained suggest that heparin alone as well as soluble proteinoid and heparin (both in water, dosed orally or by ID injection) did not appear to be absorbed from the GI tract in amounts sufficient to increase APTT values (Figure 21). Heparin in citric acid elicited some increase in APTT values, but only when dosed directly into the duodenum.
Heparin proteinoid carriers gave the highest APTT values, indicated increased absorption of heparin when dosed orally, as well as when directly injected into the duodenum
(Figures 22 and 23). While the observed activity was lower than observed with heparin proteinoid carriers (Figure 23), heparin-spiked empty proteinoid carriers showed increased APTTs over baselines. Both types of proteinoid carriers showed a much greater increase in APTT values than that observed with citric acid/heparin.
The results obtained in this Example suggest that, in the proteinoid system, proteinoid carriers are necessary for the observed increase in heparin absorption, as soluble proteinoid did not show detectable activity within the experimental limits.
Example 24: Preparation and Evaluation of M1-containing
Proteinoid carrier In this Example, influenza virus antigen-containing proteinoid carriers were prepared and evaluated in rats.
Preparation of M1 Proteinoid carriers
Encapsulation of M1 in proteinoid carriers was performed in the same manner described in Example 13. M1 protein, a major internal component of influenza virus, was obtained by purification of a swine influenza vaccine donated by Drug Directorate, Health Protection Branch, Bureau of Biologies, Ottawa, Ontrario Cannada. The vaccine was prepared with the high-yielding recombinant strain X-53Aa, which derives its HA and NA from the parent strain A/NJ/11/76 (H1N1) and its internal proteins, including M1, from the parent strain A/PR/8/34 (R.B. Couc et al. (1983) Ann. Rev. Microbiol., Vol. 37, pages 529-549 and B.R. Murphy (1982) Infec. Immun., Vol. 36, pages 1102-1108). M1 was purified as described by Khan et al ( (1982) J.Clin.Microbiol., Vol. 16, pages 813-820). M1 proteinoid carriers were prepared, by mixing (at 40°C), equivolumes of an aqueous solution of 100mg/ml of Glu/Asp/Tyr/Phe proteinoid in deionized water and a 10mg/mL solution of M1 protein in 1.7N citric acid and 5% gum arabic (pH 2.0). The final M1 concentration in the suspension was 1.0mg/ml.
Preparation of HA-NA-containing Proteinoid carriers and unencapsulated antigens
HA-NA antiger was isolated according to the procedure of Gallagher et al. ((1984) J. Clin.Microbiol., Vol. 20, pages 80-93). Influenza virus (A/PR8/34) was centrifuged at 90,000 G for 60 min. The viral pellets was solubilized with 0.05M acetate buffer (pH 7.0) containing 7.5% octylglucoside and re-centrifuged under the same conditions. The resulting supernatant contained approximately 90% HA and 10% NA as determined by SDS-PAGE.
HA-NA proteinoid carriers were prepared following the same protocol as for the M1 proteinoid carriers but substituted M1 for HA-NA. The final concentration of HA-NA in the suspension was also 1.0 mg/ml.
"Empty" proteinoid carriers were prepared following the sampe procedure described for the M1 proteinoid carriers, with the only modification being that a 1.7 N citric acid/gum solution was used in place of the M1/citric acid/gum solution.
Unencapsulated antigens, M1 and HA-NA, was diluted in 1.7 N citric acid, 10 mg/ml gum arabic to the same final 1mg/ml concentration.
Experimental procedure
Male Spraque Dawley rats (about 350g weight) were used in this experiment. Oral dosage was by gavage. Four groups of five rats each (the subcutaneous control group had 4) were dosed as follows: Group 1 was dosed orally with 1mg of M1 proteinoid carriers per rat (1 ml), Group 2 was dosed orally with 1 mg per rat of "empty" proteinoid carrier, Group 3 was dosed with 1 mg of unencapsulated M1 per rat of "empty" carrier, Group 3 was dosed with 1 mg of unencapsulated M1 per rat in 1 ml and Group 4 was dosed subcutaneously (SC) with 25 ug per rat of M1 in 0.3 ml. Blood samples (300 ul) were taken from each rat by tail bleeeding before dosing and at 1, 2, 3 and 4 hours post-dose (to assay for antigen) and at 14, 28, and 42 days post-dose (for antibody assay). Solutions for subcutaneous control-M1 in TRIS (no SDS) was diluted to a concentration of 167 ug/mL. An equal amount of Freunds Complete Adjuvant (FCA, Sigma) was added and the mixture was thoroughly homogenized. The final concentration of M1 in the mixtue was 83.3 ug/ml. HA-NA solutions for subcutaneous administration were prepared in the same manner except that phosphate buffered saline replaced TRIS-SDS buffer.
The same immunization and bleeding schedule was follwed when dosing with HA-NA proteinoid carrier, with the following modifications: all rats received an oral booster with HA-NA proteinoid carrier (250 ug/rat) 42 days after the first oral dose and blood samples were again taken 14 days after the booster dose. Serum derived from the samples were stored at -20ºC until assayed.
Serum anti-M1 and anti-HA-NA specific IgGs were assayed by an ELISA method as described Khan et al. ((1982) J. Clin. Microbiol., vol. 16, pages 813-820).
Results and Discussion
Attempts to measure antigen in plasma samples were unsuccessful. M1 antigen could not be detected in rat plasma samples taken 1-4 hours post-dosing in all groups, including the subcutaneous control.
Plasma samples from rats dosed orally with "empty" proteinoid carriers showed no significant antibody titer against either M1 or HA-NA antigens when assayed by ELISA (Table 8). As expected, rats dosed with 25 ug of either M1 or HA-NA antigen
(with FCA) subcutanouesly developed a vigorous antibody response with titers that ranged from 54,000-330,000 in the case of M1 and 176,750-909,000 in the case of HA-NA (Table 8).
Plasma samples from three of the five rats dosed with M1 proteinoid carriers showed a signficant primary response to M1 antigen. All three rats had titers ranging from 760 to 2150 as early as 14 days post-dosing, compared to <30 in all rats that received the amount of unencapsulated M1 (Table 8). Titers in the group that received proteinoid carriers increased to 1150-5200 by 42 days (Figure 24).
Four out of six rats immunized with unencapsulated HA-NA did show a moderate anti-HA-NA IgG response, with titers of 3400-17,675, while two of six rats dosed with HA-NA proteinoid carrier showed a significant response (Figure 25). The rats that did respond, however, reached titers at least eight times higher than those obtained in the controls. Although several rats showed higher titers after the oral booster with HA-NA proteinoid carriers given 42 days post-dose, most did not show a significant increase in titers.
The results support that a single dose of M1 proteinoid carriers was capable of inducing a significant IgG response to M1 as early as two weeks post-dosing, while rats dosed with same M1 (no proteinoid carriers) total dose showed no detectable antibody response. Similarly, a single dose of HA-NA proteinoid carriers induced a response in 33% of the rats used in the study. This response was up to eight times greater than rats dosed with unencapsulated HA-NA.
TABLE 8
ANTI M PROTEIN ANTIBODY TITERS IN SERUM FROM RATS DOSED WITH M PROTEINOID CARRIERS VS CONTROLS
14 day 28 day 42 day
Dosing rat # titer titer titer oral M protein 197 <30 <30 <30 unencapsulated 198 <30 <30 <30 1 mg/rat 199 <30 <30 <30
200 <30 <30 35
201 <30 <30 56 empty carrier 203 <30 <30 82
204 <30 <30 70
205 <30 <30 60
206 <30 <30 86
207 <30 <30 45
M proteinoid 209 <30 <30 64 carriers 210 2,150 820 5,200
1 mg/rat total 211 860 430 1,150
212 760 1,850 3,000
213 <30 <30 62 subcut. control 215 40,000 62,000 330,000 0.025 mg/rat 217 34 8,000 54,000 in FCA 218 430 8,000 125,000
219 270 6,600 78,000
Appendix A
PROTEINOID BATCHES p 1 of 9
B t. No . #AA COHPOSITION ADDITIVE TEMP TIME SPHERE BATCH SIZE Operator Date
C (hr) RATING molar
085 3 GLU2 ASP2 ILEU - - - 170 3.0 INS5 HT1 0.0
086 3 GLU2 ASP2 VAL - - - 170 3.0 INS4 HT0 HEP0 0.0
087 3 GLU ASP LEU - - - 170 3.0 1NS5 HT3 HEP5 0.0
088 2 GLU2 ASP2 EQU SEE HEH0 0.0 0.0
089 2 GLU2 ASP2 EQU - - - 170 3.0 INS5 HT0 0.0
090 3 GLU2 ASP2 VAL - - - 170 3.0 INS3 HT0 HEP1 0.0
091 3 GLU ASP LEU - - - 170 3.0 INS2 HT1 0.0
092 3 GLU ASP THR - - - 170 3.0 INS2 HT0 0.0
093 4 GLU2 ASP2 VAL PRO - - - 170 3.0 INS2 HT2 0.0
094 3 GLU ASP CYS-H - - - 170 3.0 INS1 HT1 0.0
095 4 PRO SER THR CYS - - - 170 3.0 0.0
096 3 GLU ASP VAL2 - - - 170 3.0 INS3 KTO HEP4 0.0
097 3 GLU ASP VAL - - - 170 3.0 INS2 KT1 0.0
098 3 GLU ASP CYS-H - - - 170 3.0 INS4 HT1 0.0
099 2 GLU2 ASP2 EQU - - - 170 3.0 INS4 0.0
186 -cp 4 PYGLU ASP TYR PHE PA 170 4.0 INS0 HT4 HEP5 0.3
199 -cp 4 GLU ASP TYR PHE H28 100 99.0 HT0 INS0 HEP0 0.0
202A-cp 4 GLU2.4 ASP2 VAL2 GLY - - - 170 4.0 INS3 HT0 0.6
202B-cp 4 GLU2.4 ASP2 VAL2 GLY - - - 170 4.0 HTO INS3 0.6
206A-cp 4 GLU ASP-TYR PHE SULFA 175 4.5 IR54 HT4 HEP3 0.6
206B-cp 4 GLU ASP_TYR PHE SULFA 175 4.5 0.6
206C>3k 4 GLU ASP-TYR PHE SULFA 175 4.5 0.6
207A-cp 4 GLU ASP-TYR PHE SULFA 175 10.0 INS5 HT4 HEP4 2.0
207B-cp 4 GLU ASP-TYR PHE SULFA 175 10.0 HT5 INS4 HEP4 2.0
211A-cp 4 GLU ASP-VAL LYSFB SULFA 190 4.3 IHS5 HT5 HEP5 v 0.3
211B-cp 4 GLU ASP-VAL LYSFB SULFA 190 4.5 0.3
212A-cp 3 GLU2-TYR.PHE SULFA 185 5.0 INS4 HT3 HEP4 0.3
212B-cp 3 GLU2-TYR PHE SULFA 185 5.0 0.3
214 -cp 3 GLU LYSFB-ARG SULFA 180 7.0 INS0 HT0 HEP0 0.0
223 -cp 4 LYSFB2 ARG2 LEU PGLU SULFA 180 8.0 INS0 HT0 HEP2 0.3
227A-cp 2 VAL2 GLY2 SULF 180 1.5 IHS0 HT0 HEP0 0.1
227B-cp 2 VAL2 GLY2 SULFA 180 1.5 HT0 INS0 HEP0 0.1
228A-cp 3 VAL2 CLY2 PGLU SULF 180 2.5 INS0 HT0 HEP0 0.1
228B-cp 3 VAL2 GLY2 PGLU SULFA 180 2.5 HT0 INS0 HEP0 0.1
248 -cp 3 GLU ASP LEU - - - 190 4.0 INS3 HT0 HEPa 0.0
265A-cp 4 GLU ASP-TYR PHE SUL 155 4.0 INS4 HT4 HEP5 1.0
265B 4 GLO ASP -TYR PHE SOLFOLANE 155 4.0 1.0
265C .0 .0
296A-cp 4 GLU LYSH PHE ASP SUL-H 180 3.0 INS4 HT2 HEP0 0.6
296B-cp 4 GLU LYSH PHE ASP SUL-H 180 3.0 0.6
298 -cp 4 GLO ASP-TYR PHE SUL-H 190 1.5 INS1a HT3 HEP4 0.5
301 -cp 4 GLO ASP-TYR PHE SUL 175 8.0 INS4 HT2 HEP3 2.0
302 -cp 4 GLO ASP-TYR PHE HHeP0 190 1.5 INS4 HT2 HEP3 0.3
308 -cp 4 GLO ASP TYR PHE HHP 170 1.0 INS4 HT4 HEP4 0.3
309 -cp 4 -GLU1.3 ASP1.3 TYR PHE1.3 SOLFOLANE 190 1.5 INS4aMT3oaHEP4a 0.3
310 -cp 4 -GLO ASP TYR PHE SOLFA LANE 190 4.0 INS4 HT2 HEP5 1.0
Glossary: a=anorphous o=oil * =varying teaperature + =cook time change PROTEINOID BATCHES Appendix A
Page 2 of 9
Bt. No. #AA COHPOSITION ADDITIVE TEMP TIME SPHERE BATCH SIZE Operator Date
C (hr) RATING molar
038 2 GLU2 ASP2 EQU - - - 150 1.5 0.0
039 3 ASP2 ARG ILEU - - - 170 0.0 HT0 0.0
040 2 GLU2 ASP2 EQU - - - 175 3.0 0.0
041 2 GLU2 ASP2 EQU PA 170 3.0 0.0
042 2 GLU2 ASP2 EQU GLYC 170 3.0 HT0 0.0
043 2 GLU2 ASP2 EQU GLYC 170 3.0 INS4 HT4 0.0
044 2 GLU2 ASP2 EQU GLYC 170 3.0 HT0 0.0
045 2 GLU2 ASP2 EQU PA 170 3.0 HT1 0.0
046 2 GLU2 ASP2 EQU GLYC 190 6.0 HT0 0.0
047 2 GLU2 ASP2 EQU PA 190 6.0 HT0 0.0
048 2 GLU2 ASP2 EQU - - - 190 6.0 HT0 0.0
049 2 GLU2 ASP2 EQU - - - 190 3.0 HT0 0.0
050 2 GLU2 ASP2 EQU - - - 170 3.0 HT0 0.0
051 2 GLU2 ASP2 EQU - - - 170 6.0 0.0
052 2 GLU2 ASP2 EQU - - - 170 6.0 HT0 0.0
053 2 GLU2 ASP2 EQU - - - 170 4.0 INS0 HT0 0.0
054 2 GLU2 ASP2 EQU - - - 200 3.5 INS4 HT0 0.0
055 2 GLU2 ASP2 EQU - - - 150 3.5 HT-VERY SH 0.0
056 2 GLU2 ASP2 EQU - - - 110 4.3 HT0 0.0
057 2 GLU2 ASP2 EQU - - - 150 3.5 HT0 0.0
05S 2 GLU2 ASP2 EQU - - - 180 5.0 0.0
059 2 GLU2 ASP2 EQU - - - 150 3.0 INS0 HT0 0.0
060 2 GLU2 ASP2 EQU - - - 160 3.0 HT3 0.0
061 2 GLU2 ASP2 EQU - - - 165 3.0 HT & N0 AM 0.0
062 2 GLU2 LEU - - - 170 3.0 HT0 0.0
063 2 GLU2 ASP2 EQU - - - 170 3.0 0.0
064 2 GLU2 LEU - - - 170 3.0 INS2 HT0 0.0
065 3 GLU2 ASP2 LEU - - - 170 3.0 INS5 HEP0 H 0.0
066 2 GLU2 GLY - - - 170 3.0 MT0 0.0
067 2 ASP2 LEU - - - 165 3.0 0.0
068 2 ASP2 LEU - - - 0.0 0.0
069 2 GLU2 ASP2 EQU - - - 170 3.0 INS5 & AHORPEOU 0.0
070 3 GLU2 ASP2 LEU - - - 170 6.0 HE 0.0
071 3 GLU ASP3 LEU - - - 170 2.6 0.0
072 2 GLU2 ASP2 EQU - - - 170 3.0 INS0 HT0 0.0
073 3 GLU ASP PRO - - - 170 4.0 τm HT0 HEP0 0.3
074 2 GLU2 ASP2 EQU - - - 170 3.0 INS5 0.0
076 2 GLU2 ASP2 EQU - - - 170 3.0 HT3 N0 AH0 0.0
077 2 GLU2 ASP2 EQU - - - 170 4.5 INS5 0.0
078 2 GLU2 ASP2 EQU - - - 170 4.0 INS5 0.0
079 4 GLU ASP PRO LYS3 - - - 170 4.5 LOST BATCH 0.0
080 3 GLU2 ASP2 ILEU - - - 170 4.0 INS4 HT0 HEP0 0.0
081 2 ARG LYS EQU - - - 170 3.0 0.0
082 2 GLU2 ASP2 EQU - - - 170 4.0 INS4 HT3 0.0
083 3 GLU2 ASP2 ILEU - - - 170 6.0 INS4 HT1 HEP4 0.0
084 3 GLU2 ASP2 ILEU - - - 170 3.0 INS4 HT3 0.0
Glossary: a=arorphous o=oil * =varying temperature + =cook ti me change Appendix A
Page 3 0f 9 PROTEINOID BATCHES Bt. No . #AA COHPOSITION ADDITIVE TEHP TIME SPHERE BATCH SIZE Operator
C (hr) RATING molar
311-cp 4 -GLU2 LYSH2 PHE2 ASP SULFALANE 190 1.7 IND40 HT30 HEP3 1.7
312-cp 4 -GLU2 LYSH2 PHE2 ASP SULFALANE 190 0.7 INS4 HT2 EEP4a 17.9
313-cp 4 -GLU2 LYSH2 PHE2 ASP SULFALANE 180 3.0 INS3 HT3 HEP3ao 0.6
314-cp 4 -ASP TYR PHE PGLU SULFOLANE 190 2.5 INS2a HT4aHEP4a 0.6
315-cp 4 GLU ASP-VAL LYSFB sulfolane 190 4.0 INS4 HT4 HEP3 0.3
316-cp 4 GLU ASP-TYR PHE sulfolane 180 21.0 INS4 HT3a HEPa 0.3
31 7-CP 4 GLN-ASP TYR PHE SULFALANE 175 4.0 INS5 MT5 HEP5 0.3
318-cp 5 GLU2 ASP2 TYR2 PHE2 0RN sulfalane 180* .0 HT1 INS4 HEP3a 1.0
319 -cp 4 -TYR PHE ASP PGLU SULFALANE 190 2.5 INS4aHT4 HEP4a 0.3
320-CP 4 -TYR PHE PGLU ASP Sulfolane 190 1.5 INS4aHT4 HEP4 0.3
321-cp 5 GLU2 ASP2 TYR2 PHE2 ORN SULFALANE 180* 3.0 INS3aHT2aHEP4a 1.0
322-cp 4 GLU2 LYSH2 PHE2 ASP- SULFALANE 192 1.2 INS2 HT2 HEP2 0.6
323-cp 4 GLU ASP TYR PHE- SULFALANE 190 .0 ABORT 16.0
324-cp 4 -GLU ASP TYR PHE SULFALANE 190 3.0 INS4 HT4 HEP5a 2.0
325-cp 5 GLU2 ASP2 TYR2 PBE2 ORH SULFALANE 180* 3.0 INS5a HT2a HEP 1.0
326-CP 4 -GLU ASP TYR PHE SULFALANE 190 6.5 INS3a HT0a HEP3 16.0
326-CP 4 -GLU ASP TYR PHE SULFALANE .0 INS4a HT4 HEP4a .0
327-CP 4 -GLU ASP TYR PHE SULFALANE 190 4.0 INS4a HT5a BEP3 17.0
32S-CP 4 -GLU ASP TYR PHE SULFALANE 190 3.0 INS5a HT3aHEP3a 17.0
328-7e 4 -GLU ASP TYR PHE SULFALANE .0 INS3a HT0a HEP4 .0
329-cp 4 -GLN ASP TYR PHE SULFALANE 175 6.5 INS5a HT3a HEP5 1.0
330-cp 2 ASP PHE SULFALANE 180 3.0 INS0a HT1a HEPO 0.5
331-CP 2 ASP2 PHE SULFALANE 180 3.0 INS0a HT0a HEPO 0.5
332 332 2 ASP3 PHE SULFALANE 180 3.0 INS3aHT1aHEP0c 0.5
333-7a 4 -GLU ASP TYR PHE SULFALANE 190 5.0 INS2aHT4a HEP5a 17.0
334-7ov 4 -GLU ASP TYR PHE SULFALANE 190 5.0 INS4aHT5a HEP4a 17.0
335-CP 2 -ASP PHE2 SGLFOLAKI 180 3.0 INS1aHT2aHEP1a 0.5
336-11 5 -GL02 ASP2 TYR2 PBE2 ORH SULFALANE 180 .0 INS3a MT3a HEP4 2.0
337 337 2 -ASP2 TYR SOLFOLAHE 180 6.5 INS2aMT0cHEP0c 0.5
338-CP 2 -ASP TYR SULFALANE 180 3.0 INS0 MT0 HEP0 1.0
339 -CP 2 -ASP3 TYR SULFALANE 180 3.0 INS0a MT0 HEP0 0.5
340 4 -GLU ASP TYR PHE SULFALANE .0 1.5
341 4 -GLU ASP TYR PHE SULFALANE .0 17.0
342 342 2 -ASP TYR2 SULFALANE .0 INS0HT0HEP0 0.5
342 -CP 2 -ASP TYR2 SULFALANE .0 INS0a MT0 HEP0 0.5
343 4 -GLU ASP TYR PHE SULFALANE .0 17.0
344 4 -GLU ASP TYR PHE SULFALANE .0 2.0
345 -CP 2 - ASP2 PHE SULFALANE .0 INS0a HT0 HEP0 . .0
346 -CP 4 -GLH ASP TYR PHE SULFALANE .0 INS0aHT1aHEP2A .0
347 4 - GLU2 ASP2 TYR5 PHE5 SULFALANE .0 .0
348 2 -ASP2 PHE SULFALANE .0 .0
349 2 -PHE ASP2 SULFALANE .0 .0
350 2 -ASP2 PHE SULFALANE .0 .0
351 351 3 -GLU2 TYR PHE SULFALANE .0 INS3aHT2aHEP3a .0
352 5 -GLU2 ASP2 TYR2 PHE2 ORN SOLFOLAHE .0 .0
353 5 -GLU2 ASP2 TYR2 PHE2 ORN SULFALANE .0 .0
Glossary: a=amorphous o=oil * =varying temperature + =cook time change PROTEINOID BATCHES Appendix A page 4 of 9
Bt. No. #AA COMPOSITION ADDITIVE TEMP TIME SPHERE BATCH SIZE
C (hr) RATING molar
295 -CP 1 ASP SUL-H 180 1.5 INS2aHT2aHEP3oa 0.3
297-cp 4 GLU ASP-TYR PHE SUL-H 190 1.5 INS2a HT4a HEP3 0.5
299-cp 4 GLU LYS PHE ASP SUL-H 190 1.7 INS5 HT4 HEP2 0.6
300-cp 5 GLU ORN ASP LYS PHE - - - - 180 3.0 INS3 HT3 HEP3 0.3
303-cp 4 GLU ASP-TYR PHE SUL-H 175 8.0 INS4 HT2 HEP3a 2.0
304-cp 5 GLU ASP-TYR PHE ORH0.5 SUL-H 180 3.0 INS4 HT2 HEP3 2.0
305 4 -PGLU ASP.5TYR PHE SUL 0.0 INS3 HT2 HEP3 0.3
306-cp 4 -GLU ASP .5TYR PHE SUL 0.0 INS3aHT2aHEP2a 0.3
307-cp 4 GLN ASP TYR PHE SULFOLANE 175 4.0 INS4o HT4 HEP4o 0.3
- .0 .0
000 .0 .0
001 2 GLu2 ASP2 EQU - - - 170 4.0 0.0
002 2 GLU ASP EQU - - - 149 0.0 0.0
003 2 GLU ASP EQU - - - 163 0.0 0.0
004 0 - - - 204 0.0 0.0
005 2 GLU ASP EQU - - - 176 3.0 0.0
006 2 GLU ASP EQU - - - 154 3.0 0.0
007 2 GLU ASP EQU - - - 196 2.0 0.0
008 2 GLU ASP EQU - - - 154 3.6 0.0
009 2 GLU2 ASP2 EQU - - - 192 3.0 0.0
010 2 GLU2 ASP2 EQU - - - 163 4.0 0.0
011 2 GLU2 ASP2 EQU - - - 160 5.0 0.0
012 2 GLU2 ASP2 EQU - - - 154 4.0 0.0
013 2 GLU2 ASP2 EQU - - - 176 4.0 0.0
014 2 GLU2 ASP2 EQU - - - 174 3.5 0.0
016 2 GLU2 ASP2 EQU - - - 170 3.5 0.0
017 2 GLU2 ASP2 EQU - - - 170 3.5 0.0
018 2 GLU2 ASP2 EQU - - - 170 3.5 0.0
019 2 GLU ASP EQU - - - 180 3.5 0.0
020 2 GLU2 ASP2 EQU - - - 180 4.5 HT 0.0
021 2 GLU2 ASP2 EQU - - - 180 3.5 0.0
022 2 GLU2 ASP2 EQU - - - 180 3.5 0.0
023 2 GLU2 ASP2 EQU - - - 180 3.3 0.0
024 2 GLU2 ASP2 EQU - - - 175 3.3 0.0
025 2 GLU2 ASP2 EQU - - - 175 3.0 0.0
026 3 GLU2 ASP2 ASPG - - - 175 3.0 0.0
027 3 GLU2 ASP2 SER - - - 195 5.0 0.0
028 2 GLU2 ASP2 EQU - - - 175 3.5 0.0
029 2 GLU2 ASP2 EQU - - - 175 3.5 0.0
031 2 GLU2 ASP2 EQU - - - 170 3.3 0.0
032 2 GLU2 ASP2 EQU - - - 170 3.5 HT0 0.0
033 2 GLU2 ASP2 EQU - - - 175 3.0 HT0 0.0
034 2 GLU2 ASP2 EQU - - - 180 0.0 HT0 0.0
035 2 GLU2 ASP2 EQU - - - 3.0 HT0 0.0
036 2 GLU2 ASP2 EQU - - - 175 3.6 HT0 0.0
037 2 GLU2 ASP2 EQU - - - 175 21.0 0.0
Glossary: a=amorphous o=oil * =varying temperature + =cook time change Appendi x A
PROTEINOID BATCHES page 5 of 9
Bt. No. #AA COMPOSITION ADDITIVE TEMP TIHE SPHERE BATCH SIZE
C (hr) RATING molar
249 <3K 4 GLU2LEU2LYSH2PGLU - - - 180 3.0 INS0 HT0 HEP0 0.1
250 <3K 5 PGLUARGH2LYS2LEUASP2 - - - 180 3.0 INS0 HT0 HEP0 0.1
251 <3K 4 GLU2ASP2TYR5-PHE5 SUL-H 180 3.0 INS4 HT4 HEP2 0.1
252 -cp 4 (GLU+ASP)VAL LYS - - - 170 3.0 INS1 HT2 HEP1 0.0
253 -cp 4 GLU ASP-TYR PHE SUL-H 180 4.5 INS1 HT0 HEP0 2.0
253 4 GLU ASP-TYR PHE SUL-H 180 10.0 INS4 HT4 HEP4 1.0
254 -cp 5 GLU2ASP2-TYR2PHE2ORN SUL-H 180 8.5 INS4 HT4 HEP4 0.1
255 -cp 4 GLU ASPTYR-PHE ORN SUL-H 180 3.0 INS2 HT4 HEP4 0.3
256 -cp 4 GLU2LYSH2PHE2PGLU - - - 180 3.0 INS0 HT0 HEP0 .0.1
257 -cp 4 GLU ASP ARGH ORNH - - - 180 3.0 INS0 HT0 HEP0 0.0
258 <3K 3 GLU ASP ARGH - - - 180 3.0 INS1 HT1HEP0 0.0
259 <3K 4 GLU ASP-TYR PHE SUL-H 180 3.0 INS3 HT3 HEP3 0.3
260 <3K 4 GLU ASP-TYR PHE SUL-H 180 2.5 INS2 HT3 HEP2 0.3
261-cp 4 GLU ASP-TYR PHE SUL-H 180 3.0 INS0 HT0 HEPO 0.3
262-cp 4 GLU LORN ASP LYSFB SUL-H 180 3.5 INS0 HT1 HEP4 0.3
263-cp 4 GLU2 LYSH2 PHE2 ASP - - - 190 3.0 INS3 HT3 HEP0 0.3
264-cp 4 GLU2 LYSH2 PHE2 ASP - - - 180 3.2 INS5 HT3 HEP4 0.3
266-cp 4 GLU2 LYSH2 PHE2 ASP - - - 180 3.0 INS4 HT4 HEP4 0.3
267-cp 3 GLU LYSFB ASP LYSFB SUL-H 180* 3.0 INSa HTc HEPc 0.3
268 4 GLU ASP-TYR PHE SUL-H 190 2.5 INS0 HT0 HEP0 0.3
269-cp 4 GLU ORHH ASP-LYSFB SUL-H 180 4.0 INSc HTc HEPc 0.1
270-cp 4 GLU ASP-TYR PHE SUL-H 180 1.5 INS5 HT4 HEP0 1.5
271 3 GLU LYSFB-PHE SUL-H 190 1.5 INS3aHT4oHEP4o 0.0
272-cp 4 GLU2 LE02 LYSH2 TYR1 - - - 180 3.0 INSC HT1 HEP4 0.1
273-cp 4 GLU2 LE02 LYSH2 PHE1 - - - 180 3.0 INS2aHT2 HEP2,a 0.1
274-cp 4 GLU LEO ARG TYR - - - 180 3.0 INSc HTc HEPc 0.1
275-cp 4 GLU ARGH-TYR SUL 190 1.5 INSc HTc HEPc 0.3
276-cp 4 GLU2 LE02 ARG2 PHE - - - 180 3.0 INS3 HT3 HEP4 0.1
277-cp 3 GLU LYS TYR SUL-H 190 1.5 INSc HTc HEP4o 0.3
278-cp 3 GLU LYS PHE SUL-H 190 1.5 INSc KTc HEP4 0.3
279-cp 3 GLU LYS ALA - - - 190 1.5 INSc HTc HEPc 0.3
280-cp 4 GLUGLUASPGLUTYRGLPHE SUL-H 190 1.5 INS4 HT3 HEP4 0.4
281-cp 4 GLU1 ASP1 TYR2.5-PHE2.5 SUL-H 180 3.0 INS4 HTa HEP2a 1.0
282-cp 3 GLU2 LYS5 PHE2 - - - 190 1.5 INS0 HT0 HEP2 0.3
283-cp 4 GLU2 LYS5 PHE5 TYR2 - - - 190 1.5 INS0 HT0 HEP3 0.1
284 5 GLU2ASP2-TYR2PHE20RN SUL-H 180 3.0 INS4aHT4oHEP2a 1.0
285 -cp 2 GLU(2X) ASP(2X) - - - 180 3.0 INSc HTc HEPc 0.3
286 -cp 2 GLU ASP(2X) - - - 180 2.5 INSc HTc HEPc 0.3
287 -cp 2 GLU PHE - - - 180 3.5 INS3 HT2HEP3 0.3
288 -cp 3 GLU ORN PHE - - - 180 3.0 INSc HTc HEPc 0.3
289 2 GLU ARG - - - 180 1.0 0.3
290-cp 3 GLU ARG PHE - - - 180 3.0 INS2 HT2 HEP2 0.3
291-CP 3 GLU LYS PHE SUL-H 190 1.5 INS4 HT3o HEP4o 0.3
292-cp 5 GLU ASP ARG ORN PHE SUL-H 180 3.0 INS0 HT0 HEP0 0.3
293-cp 4 GLU ASP ARG ORN PHE SUL-H 180 3.0 INS3 HT3 HEP3 0.3
294-cp 4 GLU2 LYSH2 PHE2 ASP 180 3.0 INS3 HT4 HEP4 0.3
Glossary: a=amorphous o=oil * = varying temperature + =cook ti me change Appendix A
Page 6 of 9
PROTEINOID BATCHES Bt. No. #AA COHPOSITION ADDITIVE TEMP TIME SPHERE BATCH SIZE
C (hr) RATING molar
192 -cp 3 GLU LYSFB ASP - - - 195 3.0 INS4 HT0 0.3
193 >6K 4 (GLU+ASP) TYR PHE PA 175 4.0 HT0 HEP0 0.3
194 -cp 3 GLU LYSFB ASP TRIGL 195 3.0 INS1 HT0 HEP2 0.3
195 -cp 3 GLU ASP VAL2 - - - 170 3.2 INS2 MT1 HEP0 0.3
196 -cp 4 GLU ASP TYR PHE PA 175 4.2 INS2 KT5 HEP4 1.0
197 -cp 4 GLU ASP TYR PHE SOL 175 2.7 INS2 HT5 HEP5 0.3
198 -cp 3 GLU LYSFB ASP - - - 195 3.2 INS3 1.0
200 -cp 3 GLU LYSH ASP PA 185 3.0 INS4 HT0 HEP0 0.3
201-cp 3 GLU LYSFB ASP SULPA 195 3.0 INS4 HT0 HEP3 0.3
203-cp 4 GLU ASP VAL LYS - - - 170 3.0 INS5 HT5 HEP 0.3
204-cp 3 GLU LYS ASP - - - - - 185 3.0 INS4 HT0 HEP0 0.3
205 -cp 4 GLU ASP-TYR PHE SULFA 175 3.7 INS4 HT0 0.6
208 3 GLUH LYSH ASPH NaHCO&MeOH 80 8.0 INS0 HT0 HEP0 0.0
209-cp 4 GLU ASP-VAL LYS SULFA 170 3.2 INS4 HT4 HEP3 W 0.3
210-cp 4 GLU ASP-VAL LYSFB SULFA 170 3.0 INS4 HT4 HEP3 2.0
213-cp 3 GLU-LYS HIS SULFA 180 3.0 INS0 HT0 HEP0 0.3
215-cp 3 GLU ASP GLY2 - - - 180 5.5 INS3 HT0 HEP0 0.3
216-cp 4 GLU ASP-TYR PHE SULFA 175 3.0 INS4 HT4 HEP4 2.0
217 3 GLUASPLYS(DIETESTER) HE0H/Et3N 75 29.0 0.0
218-cp 4 GLU ASP-TYR PEE SOLFA 175 .0 INS0 HT0 HEP0 A 0.3
219-cp 3 GLU-LYS-LEU Sul/POCl3 180* 8.5 INS2 HT0 HEP2 0.3
220-cp 4 GLU ASP-TYR PHE SULFA 180 20.5 INS4 HT4 HEP5 0.3
221-cp 3 -ASP2 TYR PHE SULFA 180 22.0 INS2 HT0 HEP0 0.3
222-cp 3 -LYSFB2 ARG2 LEU SULFA 180 4.0 INS0 HT0 HEP2 0.3
224-cp 4 GLU ASP-TYR PHE- SU/PA 180 6.0 INS3 HT0 HEP0 0.3
225-cp 3 PRO-SER TYR SULF 180 3.5 INS2 HT0 HEP0 0.3
226 <3K 4 GLU ASP TYR PHE SULF 180 4.0 INS3 HT4 HEP3 0.3
229 <3K 4 -GLU ASP TYR PHE SULF 180 5.5 INS3 HT0 HEP0 0.3
230 -cp 2 GLU TYR - - - 180 4.0 INS4 HT0 HEP0 0.3
231-cp 3 GLU LYSFB PHE SULF 180 3.5 INS2 HT0 HEP0 0.3
232 -cp 3 GLU LEU ARG - - - 180 4.0 INSO HT0 HEP0 0.3
233-cp 3 GLU LEU LYSH - - - 180 4.0 INSO HT0 HEP0 0.3
234-cp 4 -(GLU ASP TYR PHE) SULF 150 27.0 INS3 HT0 HEP0 0.3
235-cp 4 -(GLU ASP)TYR10PHE10 SULF 180 22.0 INSO HT0 HEP0 0.0
236-cp 3 GLU TYR LYSHCL - - - 180 2.0 INSO HT0 HEP0 0.3
237 <3K 4 GLU2 LEU2 LYSH2 ASP - - - 180 3.0 INSO HT0 HEP0 0.1
238 <3K 4 GLU ASP TYR5 PHE5 SULF 180 3.0 INSO HT0 HEP0 0.1
239-cp 3 -GLU ASP LEU SUL-H 190 1.0 HS3 HT0 HEP0 0.0
240-cp 3 -(GLU ASP) LEU SUL-H 170 4.0 INS4 HT0 HEP0 0.0
241 <3K 3 -(GLU ASP) LEU SUL-H 190 5.0 INS3 HT0 HEP0 0.0
242-cp 3 (GLU ASP LEU) SUL-H 170 2.5 INS0 HT0 HEP0 0.0
243 -cp 5 PGLU2ASPARG2LYS2LE0 - - - 180 3.0 INS0 HT0 HEP0 0.1
244 -cp 3 (GLU ASP) LEU - - - 190 2.5 INS0 HT0 HEP0 0.0
245-cp 3 (GLU ASP) LEU - - - 170 1.0 INS3 HT1 HEP0 0.0
246 <3K 5 GL02 LYSH2 PHE2 ASP - - - 180 6.0 INS4 HT4 HEP4 0.0
247-cp 3 GLU ASP LEU - - - 170 5.0 INS0 HT0 HEP0 0.0
Glossary: a= amorphous o=oil * =varying temperature + =cook time change Appendix A
Page 7 of 9
PROTEINOID BATCHES erator Date
Bt. No. #AA COHPOSITION ADDITIVE TEMP TIME SPHERE BATCH SIZE Op
C (hr) RATING molar
145 3 GLU ASP LYSFB PPA 185 6.0 HT0 0.0
146 3 GLU ASP VAL2 PPA 170 3.5 INS2 HT0 0.0
147 4 GLU ASP PHE ALA - - - 170 3.0 INS4 HT3 HEP4 0.0
148 4 GLU ASP TYR PHE PA 170 3.0 HT0 HEP4 0.0
149 3 GLU ASP PHE2 - - - 170 3.0 0.0
150 4 GLU ASP LEU PHE PA 170 24.0 HT0 HEP0 0.0
151 4 GLU ASP TYR PHE PA 170 6.0 INS4 HT4 HEP4 0.0
152 4 GLU ASP TYR PHE PA 170 5.0 0.0
153 3 GLU LYSEB PHE PA 170 24.0 0.0
154 4 GLU ASP TYR HE PA 170 4.0 0.0
155 3 GLU2TYRPHE PA 170 4.0 INS4 KB HEP3 0.0
156 3 GLU4 LYS2 PHE - - - 170 6.0 INS0 HT0 HEPc 0.0
157 3 GLU2 TYR LEU PA 170 5.0 INS2 HT1 HEP0 0.0
158 3 GLU2 PHE LEU PA 175 5.0 INS4 HT0 HEP4 0.0
159 3 GLU3 PHE TYR PA 175 5.0 INS4 HT4 HEP4 0.0
Tc HEPc 0.0
160 4 GLU6 LYS2 PHE TYR PA 170 6.0 INSa H
161 4 GLU4 PHE2 TYR2 CYS PA 170 4.0 INS4 HT HEP 0.0
162 3 GLU2 TYR PHE PA 170 5.5 INS3 HT0 HEP2 0.0
163 3 GLU2 PHE TYR PA 170 5.0 INS3 HT2 HEP3 0.0
164 3 GLU2 PHE TYR PA 170 5.0 INS4 HT4 HEP4 0.0
165 4 GLU3 ASP PHE2 TYR2 PA 170 3.0 INS3 HT0 HEP0 0.0
166 3 GLU LYSFB PGLU PA 170 7.0 0.0
167 4 GLU ASP TYR PHE PA 170 6.5 0.0
168 3 GLU ASP LYSFB PPA 185* 72.0 HT0 0.0
169 3 GLU ASP LYSFB PPA 185 72.0 HT0 0.0
170 3 GLU ASP LYSFB - - - 195 7.0 HT5 0.0
171 3 GLU LSYHCL ASP H.OIL 180 7.0 HT0 0.0
172 4 GLU ASP TYR PHE PA 170 6.0 HT1 0.0
173 3 GLU LYS ASP nineral o .185 3.0 ABORT 0.3
P3 0.3
174 >6K 3 GLU LYS ASP GLycerin 185 3.0 INS2 HT1 HE
175 >6K 4 GLU ASP TYR PHE PA 172 3.5 1.0
176 >6K 3 GLU2 LYS2 LYS - - - 180. 3.0 INS0 HT0 HEP0 0.3
177 >6K 3 GLU ARG ASP - - - 180 3.2 INS0 HT2 HEP0 0.3
178 >6K 3 GLU LYS ASP - - - 190 3.2 INS0 HT0 HEP1 0.3
179 >6K 4 GLU ASP TYR PHE PA 175 4.0 1.0
180 >6K 4 GLU ASP TYR PHE PA 175 7.0 See Hotes. 1.0
181 >6K 3 GLU LYS ASP - - - 185 3.0 INS0 HT0 0.3
182 >6K 4 GLU ASP TYR PHE PA 175 3.7 HT1 HEP1 0.3
183 4 PGLU ASP TYR PHE PA 175 4.0 ABORT-RETRY 0.3
184 -cp 4 GLU ASP TYR PHE PA 175 3.5 HT2 HEP4 0.3
185 -cp 4 GLU ASP TYR PHE PA 176 4.2 INS HT4 HEP4 1.0
187 3 ASP TYR PHE PA 170 .0 ABORT 0.3
188-cp 3 ASP TYR PHE PA 150 21.2 INS0 HT0 HEP0 0.3
189-cp 4 GLU ASP TYR PHE PA 176 4.0 HT4 HEP5 1.0
HEP4 1.0
190-cp 4 GLU ASP TYR PHE PA 175 4.0 HT5
191-cp 3 ASP2 TYR PHE PA 150 24.0 HT0 HEP0 0.3
Glossary: a=amorphous o=oil * =varying tesperature + =cook the change Aopendix A
Page 8 of 9
PROTEINOID BATCHES
Bt. No. #AA COMPOSITION ADDITIVE TEMP TIME SPHERE BATCH SIZE
C (hr) RATING molar
019 2 GLU2 ASP2 EQU - - - 170 2.5 0.0
100 3 GLU ASP VAL2 - - - 170 3.0 0.0
101 3 GLU ASP VAL2 - - - 170 3.0 0.0
102 3 GLU ASP VAL2 - - - 170 3.0 0.0
103 4 GLU ASP GLY VAL - - - 170 3.5 INS4 0.0
104 4 GLU ASP VAL LEU - - - 170 3.5 INS4 HT2 HEP5 0.0
105 3 GLU ASP GLY2 - - - 180 4.0 INS4 HT2 0.0
106 4 GLU ASP VAL LEU - - - 170 5.0 0.0
107 4 GLU2 ASP2 GLY VAL2 - - - 170 3.0 INS5 0.0
108 4 GLU2 ASP2 GLY VAL2 - - - 170 4.0 INS4 H0 AHORPHO 0.0
109 4 GLU2 ASP2 GLY VAL2 - - - 170 4.0 INS4 HT1 0.0
110 4 GLU2 ASP2 GLY2 VAL - - - 170 3.5 0.0
111 5 GLU ASP GLY VAL CYS - - - 170 3.0 0.0
112 4 GLU ASP GLY PHE - - - 170 4.0 INS4 HT3 HEP4 0.0
113 4 GLU ASP VAL2 GLY - - - 170 3.0 INS2 HT0 0.0
114 3 GLU ASP VAL - - - 170 3.0 INS4 HT0 0.0
115 3 GLU VAL TYR - - - 170 4.0 0.0
116 4 GLU ASP VAL LYS - - - 170 4.0 0.0
117 3 GLU VAL TYR - - - 170 3.0 INS5 0.0
118 2 GLU TYR - - - 170 3.5 INS5 HT0 HEP0 0.0
119 2 GLU2 ASP2 EQU - - - 170 3.5 INS5 HT1 0.0
120 3 GLU ASP TYR - - - 170 4.5 INS0 HT0 HEP0 0.0
121 4 GLU ASP TYR PHE - - - 170 4.0 INS5 HT3 HEP4 0.0
122 4 GLU ASP VAL TYR - - - 170 3.0 INS3 HT0 HEP0 0.0
123 1 GLU - - - 170 4.5 CAN'T DRY 0.0
124 3 GLU TYR VAL - - - 170 3.5 INS4 HT3 HEP3 0.0
125 3 PGLU VAL TYR - - - 170 3.5 INS3 HT2 0.0
126 4 GLU ASP VAL2 GLY - - - 170 4.0 INS1 0.0
127 4 GLU2 ASP2 VAL2 PHE - - - 170 4.0 INS3 HT2 HEP4 0.0
128 2 GLU2 ASP2 EQU - - - 170 3.5 HT0 0.0
129 2 GLU2 ASP2 EQU - - - VARY 4.0 INS3 HT0 0.0
130 2 GLU2 ASP2 EQU - - - 220 3.0 INS5 HT0 0.0
131 2 GLU2 LYSFB - - - 185 3.0 INS1 HT0 0.0
132 3 GLU ASP LYSFB - - - 185 3.0 INS3 HT2 HEP2 0.0
133 3 GLU ASP LYSFB PA 180 6.2 INS5 HT1 HEP2 0.0
134 4 GLU ASP LYS VAL PA 185 3.0 0.0
135 3 GLU ASP LYSFB GLYC 185 6.5 INS1 HT1 0.0
136 2 GLU2 ASP2 EQU - - - 155 3.0 HT0 0.0
137 5 GLU2ASP2LEU THY VAL - - - 185 4.0 INS0 HT0 0.0
138 4 GLU ASP VAL TYR - - - 185 4.5 INS1 HT3 0.0
139 4 GLU ASP VAL TYR PPA 160* 72.0 INS2 HT3 0.0
140 3 GLU ASP LYSFB PPA 120* 72.0 INS5 HT2 HEP4 0.0
141 3 GLU LYSFBSYHPEPagqp - - - 185 6.0 0.0
142 3 GLU ASP LYSFB PPA 120* 72.0 INS1 HT1 0.0
143 3 GLU VAL TYR - - - 170 3.0 INS2 HT2 0.0
144 4 GLU2 ASP2 GLY VAL2 - - - 170 3.0 INS1 HT1 0.0
Glossary: a=amorphous o=oil * =varying temperature + =cook t ime change Appendix A page 9 of 9
PROTEINOID BATCHES
Bt. No. #AA COMPOSITION ADDITIVE TEMP TIME SPHERE BATCH SIZE
C (hr) RATING molar
354 5 -(GLU ASP TYR PHE)2 ORN SULPOLANE .0 .0
Sphere rating : . 0= worst 5 = best
INT = insulin
MT = empty microsphere
HEP = heparin
Sul-M = su lfolane , medical grade
Sulfa = Sulfa = Sul = Sul folane
PA = phosphoric acid
Equ = equxients
GLYC = glycerol
TRIGL = triglyme
PPA = polyphosphoric acid
M. Oil = mineral o . = mineral oil
Glossary: amorphous o=oil * =varying temperature + =cook time change
Figure imgf000068_0001
Figure imgf000069_0001
Appendix C
Sphere Testing of Externally Prepared Proteinoids
Page 1 of 3 - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -
SOI No. Compostion pH 0.85 CA 5-1 AA 0.85 α 5% AA INS/CA IHS/AA HEP/CA
+GH +GH GH/GL/CD GH/GL/CD + GH - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP001F20B1SA0º GLU ASP TYR PHE - - - - Rating 1-2 1-2 3 2-3 2-3 0 2-3
Desc. a a - - - - a a a a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP001F21B1SA07 GLU ASP TYR PHE - - - - Rating 2-3 2-3 3-4 3-4 2-3 4 4-5
Desc. a,ag a,ag a a a,p a,ag a,p pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP001F21B1SA07 GLU ASP TYR PHE - - - - Rating 3 3 4-5 3 3-4 3-4 4-5
Desc. ag,a a a ag,p ag,p ag,p a, p pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EKIP001F22B1SA7 GLU ASP TYR PHE - - - - Rating 2-3 2-3 3 4-5 0 3-4 4-5
Desc. - - - - a - - - - a,o a 0 a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP011F23B1SA8 GLU ASP TYR PHE ORH - - - - Rating 2-3 3 4 4 0-1 4 3-4
Desc. a,p a - - - - - - - - a a,o a,o pB - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP011F24SA7 GLU ASP TYR PHE ORH - - - - Rating 2 2 2-3 3 4 0-1 5
Desc. a a 0 - - - - a a,o P pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP001F25B1SA2A GLU ASP TYR PHE - - - - Rating 0-1 0 3-4 0 3-4 4-5 4-5
Desc. - - - - a,p a a a a a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP001F25B1SA5 GLU ASP TYR PHE - - - - Rating 0-1 1 2-3 2-3 0-1 3-4 2-3
Desc. a a a - - - - a a,o a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP001F26B1SA2A GLU ASP TYR PHE - - - - Rating 2-3 2-3 3 3-4 2 2-3 3-4
Desc. - - - - a - - - - - - - - a - - - - 0 pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
Rating
Desc.
pH
CA= citric acid INS= insulin
GM= gum acacia HEP = heparin
GL= gelatin CD = cyclodextrin
a= amorphous o = oil
p= particulate ag= aggregate
0 = worst 5=best Appendix C
Sphere Testing of Externally Prepared Proteinoids page 2 of 3 - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -
SOI No. Coupostion pH 0.85 CA 5% AA 0.85 CA 5% AA INS/CA INS/AA HEP/CA
+GH +GH GH/GL/CD GH/GL/CD + GM - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B02 GLU ASP TYR PHE - - - - Rating 2-3 3 5 4-5 4-5 2-3
Desc. a a a - - - - a a
pH - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B02 GLU ASP TYR PHE - - - - Rating 2 4-5 5 5 5 5 5
Desc. - - - - a - - - - - - - - - - - - - - - - pH - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B02 GLU ASP TYR PHE - - - - Rating 2-3 3 5 4-5 4-5 2-3
Desc. a a a - - - - a a
pB - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B03 GLU ASP TYR PHE - - - - Rating 2 2 3-4 3-4 5 3 3-4
Desc. a a a a a a a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
93CTAP001F014B03 GLU ASP TYR PHE - - - - Rating 0-1 0-1 5 5 5 2-3
Desc. a,ag a,ag a,ag a,ag a,ag a
- - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B03 GLU ASP TYR PHE - - - - Rating 2-3 2-3 5 5 5 3 5
Desc. ag ag a,ag - - - - a,ag,o ag,a
pH - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B03 GLU ASP TYR PHE - - - - Rating 2 2 3-4 3-4 5 3 3-4
Desc. a a a a a a a pB - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B04 GLU ASP TYR PHE - - - - Rating 0-1 0-1 4 5 2-3 0 5
Desc. a a a a a a a pB - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B05 GLU ASP TYR PHE - - - - Rating 3 3-4 5 4-5 4 4 4
Desc. - - - - a a - - - - a a a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B04 GLU ASP TYR PHE - - - - Rating 3 3-4 5 4-5 4 4 4
Desc. - - - - a a - - - - a a a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91ENIP011F15B1 GLU2ASP2TYR2PHE20RH - - - - Rating 2-3 3 4 3-4 0 3-4 4
Desc. a,ag ag - - - - a,o a - - - - - - - - pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91EHIP001F20B1SA07 GLU ASP TYR PHE - - - - Rating 2 2-3 3 2-3 4 3 4
Desc. ag ag - - - - - - - - a,o a,o - - - - pB - - - - - - - - - - - - - - - - - - - - - - - - - - - - Sphere Testing of Externally Prepared Proteinoids Appendix C pag e 3 of 3 - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -
SOI No. Coapostion pH 0.85 CA 5% AA 0.85 CA 5% AA INS/CA INS/AA HEP/CA
+GH +GH GH/GL/CD GH/GL/CD + GH - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -
Rating
Desc.
pH
91EH1P00IF25B1SA3a 6.52 Rating 2-3 2-3 3-4 4 3-4 4 5
Desc. ag,a ag - - - - - - - a,p 0 - - - - - - - - - - - - - - - - - - - -
91CTAP001P012B01 GLU ASP TYR PHE 7.5 Rating 2-3 2 5 5 4 5 5
Desc. a a - - - - - - - - a - - - - a - - - - - - - - - - - - - - - - - - - - - - - - - - - -
F005-B01 GLU2 LYSH2 PHE2 ASP 9.0 Rating - - - - - - - - 3-4 - - - - 4 3 3
Desc. - - - - - - - - a,o - - - - a,o a,o a,o pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTAPR001F010B01 GLU2 LYSH2 PHE2 ASP - - - - Rating 2-3 2 4 4 4 4 4
Desc. a a - - - - - - - - a a a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
F003-B01 GLU2 LYSH2 PHE2 ASP - - - - Rating - - - - - - - - 4 3-4 3 4-5 3
Desc. - - - - - - - - a o a a a ,o pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
F004-B01 GLU2 LYSH2 PHE2 ASP - - - - Rating - - - - - - - - 3-4 2-3 2-3 4 2-3
Desc. - - - - - - - - a,o a a a a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F011B01 GLU ASP TYR PHE ORN - - - - Rating - - - - - - - - - - - - - - - - - - - - - - - - - - - - Desc. - - - - - - - - - - - - - - - - - - - - - - - - - - - - pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F013B01 GLU ASP TYR PHE 7.9 Rating 2-3 2-3 4-5 4-5 4-5 4 5
Desc. a - - - - a,o a,o a,o a pH - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001F014B01 GLU ASP TYR PHE 8.0 Rating 0 0-1 4-5 4-5 4-5 3-4 4
Desc. a a a - - - - a a a pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTλP001F014B01 GLU ASP TYR PHE - - - - Rating 0-1 0-1 5 4 4 3 3-4
Desc. a a a,o a,o a,o a,o a,o pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -
91CTAP001FO14B01 GLU ASP TYR PHE - - - - Rating 2 2 5 5 5 3 5
Desc. ag ag,a ag - - - - ag,a ag,a - - - - pH - - - - - - - - - - - - - - - - - - - - - - - - - - - -

Claims

WHAT IS CLAIMED IS: 1. A proteinoid comprising a peptide polymer selected from the group consisting of:
i) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine and from at least one second monomer selected from the group consisting of glutamic acid pyroglutamic acid, glutamine, and aspartic acid; and
ii) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and from at least one third monomer selected from the group consisting of lysine, arginine, and ornithine,
said proteinoid being a microsphere or microcapsule. forming proteinoid and being soluble within a selected pH range.
2. The proteinoid of claim 1, said proteinoid having a molecular weight ranging between about 250 and about 2400.
3. The proteinoid of claim 2, said proteinoid having a molecular weight ranging between about 250 and about 400.
4. The proteinoid of claim 1, said proteinoid having between 2 to 20 amino acids.
5. The proteinoid of claim 4, said proteinoid having between 2 to 8 amino acids.
6. The proteinoid of claim 1, wherein said proteinoid is an acid-soluble proteinoid and said second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid, and said third monomer is selected from the group consisting of lysine, arginine and ornithine.
7. The proteinoid of claim 1, wherein said proteinoid is a base-soluble proteinoid and said second monomer is selected from the group of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid.
8. A proteinoid carrier comprising a proteinoid comprising
i) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and
ii) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and at least one third monomer selected from the group consisting of lysine, arginine, and ornithine,
said proteinoid being a microsphere- or microcapsule forming proteinoid and being soluble within a selected pH range.
9. The proteinoid carrier of claim 8, wherein said proteinoid carrier comprises a proteinoid microsphere.
10. The proteinoid carrier of claim 8, wherein said proteinoid carrier comprises a proteinoid microcapsule.
11. The proteinoid carrier of claim 8, wherein said proteinoid is an acid-soluble proteinoid and said second monomer is selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid and said second monomer is selected from the group of lysine, arginine and ornithine.
12. The proteinoid carrier of claim 8, wherein said Proteinoid is a base-soluble proteinoid and said second monomeris selected from the group of glutamic acid, pyroglutamic acid, glutamine and aspartic acid.
13. The proteinoid carrier of claim 8, wherein said proteinoid carrier having a diameter equal to or less than 10 microns.
14. The proteinoid carrier of claim 8, further encapsulating a cargo.
15. The proteinoid carrier of claim 14, wherein said cargo comprises a fragrance, cosmetic agent, dye, and water soluble vitamin.
16. The proteinoid carrier of claim 14, wherein said cargo is a biologically active agent.
17. The proteinoid carrier of claim 16, wherein said biologically active agent comprises an antigen, monoclonal antibody, calcitonin, erythropoietin, alpha interferon, heparin, insulin, growth hormone, atrial naturetic factor, factor IX, or interleukin-II.
18. A composition comprising a biologically active agent encapsulated within a proteinoid microsphere or microcapsule, said microsphere or microcapsule comprising a proteinoid comprising
i) peptide polymers made from at least one first monomer selected from the group consisting of tyrosine and phenylalanine and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and
ii) peptide polymers made from-at least one first monomer selected from the group consisting of tyrosine and phenylalanine; and from at least one second monomer selected from the group consisting of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid; and at least one third monomer selected from the group consisting of lysine, arginine, and ornithine,
said proteinoid being a microsphere- or microcapsule-forming proteinoid and being soluble within a selected pH range.
19. The composition of claim 18, wherein said proteinoid is an acid-soluble proteinoid and said second monomer is selected from the group of lysine, arginine and ornithine.
20. The composition of claim 18, wherein said proteinoid is a base-soluble proteinoid and said second monomer is selected from the group of glutamic acid, pyroglutamic acid, glutamine, and aspartic acid.
21. The composition of claim 18, wherein said biologically active agent comprises an antigen, monoclonal antibody, calcitonin, erythropoietin, alpha interferon, heparin, insulin, growth hormone, atrial naturetic factor, factor IX, or interleukin-II.
22. A pharmaceutical preparation comprising an oral dosage form of calcitonin.
23. The pharmaceutical preparation according to claim 22, further comprising a microsphere- or microcapsule-forming proteinoid.
24. A pharmaceutical preparation comprising an oral dosage form of a monoclonal antibody.
25. The pharmaceutical preparation according to claim 24, further comprising a microsphere- or microcapsule-forming proteinoid.
26. A pharmaceutical preparation comprising an oral dosage form of erythropoietin.
27. The pharmaceutical preparation according to claim 26, further comprising a microsphere- or microcapsule-forming proteinoid.
28. A pharmaceutical preparation comprising an oral dosage form of alpha-interferon.
29. The pharmaceutical preparation according to claim 28, further comprising a microsphere- or microcapsule-forming proteinoid.
30. A pharmaceutical preparation comprising an oral dosage form of Factor IX.
31. The pharmaceutical preparation according to claim 30, further comprising a microsphere- or microcapsule-forming proteinoid.
32. A method for delivering calcitonin to a mammal which comprises orally administering the pharmaceutical preparation according to claim 23.
33. A method for delivering erythropoietin to a mammal which comprises orally administering the pharmaceutical preparation according to claim 27.
34. A method for delivering alpha-interferon to a mammal which comprises orally administering the pharmaceutical preparation according to claim 29.
35. A method for delivering Factor IX to a mammal which comprises orally administering the pharmaceutical preparation according to claim 31.
PCT/US1993/005723 1992-06-15 1993-06-15 Proteinoid carriers and methods for preparation and use thereof WO1993025583A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
KR1019940704564A KR950701939A (en) 1992-06-15 1993-06-15 Protenoid Carrier, Method of Making and Use thereof
BR9306678A BR9306678A (en) 1992-06-15 1993-06-15 Proteinoid composition pharmacological composition methods for administering calcitonin erythropoietin alfa-interferon and ix to a mammal and method for preparing a proteinoid
JP6501793A JPH07508004A (en) 1992-06-15 1993-06-15 Proteinoid carriers and their production and use methods
AU46356/93A AU4635693A (en) 1992-06-15 1993-06-15 Proteinoid carriers and methods for preparation and use thereof
EP93916542A EP0642532A1 (en) 1992-06-15 1993-06-15 Proteinoid carriers and methods for preparation and use thereof
NO944852A NO944852L (en) 1992-06-15 1994-12-14 Proteinoid carriers and processes for their preparation and use
FI945912A FI945912A (en) 1992-06-15 1994-12-15 Proteinoid carriers and processes for their preparation and their use

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US89890992A 1992-06-15 1992-06-15
US898,909 1992-06-15
US920,346 1992-07-27
US07/920,346 US5443841A (en) 1992-06-15 1992-07-27 Proteinoid microspheres and methods for preparation and use thereof
US076,803 1993-06-14
US08/076,803 US5578323A (en) 1992-06-15 1993-06-14 Proteinoid carriers and methods for preparation and use thereof

Publications (3)

Publication Number Publication Date
WO1993025583A2 true WO1993025583A2 (en) 1993-12-23
WO1993025583A9 WO1993025583A9 (en) 1994-02-03
WO1993025583A3 WO1993025583A3 (en) 1994-08-04

Family

ID=27372957

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/005723 WO1993025583A2 (en) 1992-06-15 1993-06-15 Proteinoid carriers and methods for preparation and use thereof

Country Status (11)

Country Link
US (3) US5578323A (en)
EP (1) EP0642532A1 (en)
JP (1) JPH07508004A (en)
KR (1) KR950701939A (en)
AU (1) AU4635693A (en)
BR (1) BR9306678A (en)
CA (1) CA2138146A1 (en)
CZ (1) CZ315494A3 (en)
FI (1) FI945912A (en)
HU (1) HUT70211A (en)
WO (1) WO1993025583A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0706798A1 (en) * 1994-07-22 1996-04-17 Sanwa Kagaku Kenkyusho Co., Ltd. Pharmaceutical composition containing biologically active peptide or protein
EP0706375A1 (en) * 1993-06-14 1996-04-17 Emisphere Technologies, Inc. Proteinoid carriers
US5548064A (en) * 1993-05-24 1996-08-20 Biotech Australia Pty Limited Vitamin B12 conjugates with EPO, analogues thereof and pharmaceutical compositions
EP0831784A1 (en) * 1995-06-07 1998-04-01 Emisphere Technologies, Inc. Fragrances and flavorants
US5935601A (en) * 1994-04-22 1999-08-10 Emisphere Technologies, Inc. Modified amino acids for drug delivery
WO1999064495A1 (en) * 1998-06-05 1999-12-16 Ústav Makromolekulární Chemie Akademie Věd České Republiky FUNCTIONALISED POLYMERS OF α-AMINO ACIDS AND THE METHOD OF PREPARATION THEREOF
US6652875B1 (en) 1998-07-29 2003-11-25 Pacific Biolink Pty. Limited Casein formulations for the delivery of bioactive constituents
US7282216B2 (en) 2001-11-12 2007-10-16 Alkermes Controlled Therapeutics, Inc. Biocompatible polymer blends and uses thereof

Families Citing this family (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6331318B1 (en) 1994-09-30 2001-12-18 Emisphere Technologies Inc. Carbon-substituted diketopiperazine delivery systems
US5714167A (en) 1992-06-15 1998-02-03 Emisphere Technologies, Inc. Active agent transport systems
US5578323A (en) 1992-06-15 1996-11-26 Emisphere Technologies, Inc. Proteinoid carriers and methods for preparation and use thereof
US6221367B1 (en) * 1992-06-15 2001-04-24 Emisphere Technologies, Inc. Active agent transport systems
US6099856A (en) 1992-06-15 2000-08-08 Emisphere Technologies, Inc. Active agent transport systems
US5693338A (en) 1994-09-29 1997-12-02 Emisphere Technologies, Inc. Diketopiperazine-based delivery systems
US6916489B2 (en) * 1992-06-15 2005-07-12 Emisphere Technologies, Inc. Active agent transport systems
US5811127A (en) 1992-06-15 1998-09-22 Emisphere Technologies, Inc. Desferrioxamine oral delivery system
US5792451A (en) 1994-03-02 1998-08-11 Emisphere Technologies, Inc. Oral drug delivery compositions and methods
US5401516A (en) 1992-12-21 1995-03-28 Emisphere Technologies, Inc. Modified hydrolyzed vegetable protein microspheres and methods for preparation and use thereof
US5643957A (en) 1993-04-22 1997-07-01 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US20010003001A1 (en) 1993-04-22 2001-06-07 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5958457A (en) 1993-04-22 1999-09-28 Emisphere Technologies, Inc. Compositions for the delivery of antigens
US5713674A (en) * 1994-10-06 1998-02-03 Pfu Limited Paper feed method and apparatus for a printer
US6001347A (en) 1995-03-31 1999-12-14 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5866536A (en) 1995-03-31 1999-02-02 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6090958A (en) 1995-03-31 2000-07-18 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
CN1151836C (en) 1995-03-31 2004-06-02 艾米斯菲尔技术有限公司 Compound and compositions for delivering active agents
US5965121A (en) 1995-03-31 1999-10-12 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5989539A (en) 1995-03-31 1999-11-23 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6051258A (en) 1995-06-07 2000-04-18 Emisphere Technologies, Inc. Proteinoid emulsions and methods for preparation and use thereof
US5750147A (en) 1995-06-07 1998-05-12 Emisphere Technologies, Inc. Method of solubilizing and encapsulating itraconazole
WO1997010197A1 (en) 1995-09-11 1997-03-20 Emisphere Technologies, Inc. METHOD FOR PREPARING φ-AMINOALKANOIC ACID DERIVATIVES FROM CYCLOALKANONES
WO1997047288A1 (en) 1996-06-14 1997-12-18 Emisphere Technologies, Inc. Microencapsulated fragrances and method for preparation
CA2243643A1 (en) 1996-11-18 1998-05-28 Susan Haas Methods and compositions for inducing oral tolerance in mammals
US5876710A (en) 1997-02-07 1999-03-02 Emisphere Technologies Inc. Compounds and compositions for delivering active agents
US5939381A (en) 1997-02-07 1999-08-17 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5879681A (en) 1997-02-07 1999-03-09 Emisphere Technolgies Inc. Compounds and compositions for delivering active agents
US6060513A (en) 1997-02-07 2000-05-09 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6358504B1 (en) 1997-02-07 2002-03-19 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6313088B1 (en) 1997-02-07 2001-11-06 Emisphere Technologies, Inc. 8-[(2-hydroxy-4-methoxy benzoyl) amino]-octanoic acid compositions for delivering active agents
US5804688A (en) 1997-02-07 1998-09-08 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5990166A (en) 1997-02-07 1999-11-23 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5863944A (en) 1997-04-30 1999-01-26 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5962710A (en) 1997-05-09 1999-10-05 Emisphere Technologies, Inc. Method of preparing salicyloylamino acids
US6440929B1 (en) 1998-07-27 2002-08-27 Emisphere Technologies, Inc. Pulmonary delivery of active agents
HUP0103318A2 (en) 1998-07-27 2002-01-28 Emisphere Technologies Inc. Process for pulmonary delivery of active agents
CA2339765C (en) * 1998-08-07 2009-04-28 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6991798B1 (en) 1998-08-07 2006-01-31 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
BR0008590A (en) 1999-01-08 2001-10-30 Emisphere Tech Inc Polymeric delivery agent, composition, unit dosage form, method for administering a biologically active agent to an animal requiring the agent, method for preparing a composition; it's composed
WO2000050386A1 (en) 1999-02-26 2000-08-31 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US9006175B2 (en) 1999-06-29 2015-04-14 Mannkind Corporation Potentiation of glucose elimination
US7279597B1 (en) 1999-11-05 2007-10-09 Emisphere Technologies, Inc. Phenyl amine carboxylic acid compounds and compositions for delivering active agents
US7129274B1 (en) 1999-11-05 2006-10-31 Emisphere Technologies Inc. Phenoxy carboxylic acid compounds and compositions for delivering active agents
IL134701A0 (en) * 2000-02-23 2001-04-30 J P M E D Ltd Homogeneous solid matrix containing vegetable proteins
US6555543B2 (en) 2000-08-04 2003-04-29 Dmi Biosciences, Inc. Method of using diketopiperazines and composition containing them
US20030225300A1 (en) * 2001-04-19 2003-12-04 Emisphere Technologies Inc. Compounds and compositions for delivering active agents
AU2002365255A1 (en) * 2001-10-02 2003-09-02 The Regents Of The University Of California Nanoparticle assembled hollow spheres
US20030138975A1 (en) * 2001-12-20 2003-07-24 Kimberly-Clark Worldwide, Inc. Diagnostic signal amplification with proteinoid microspheres
US7056535B2 (en) * 2001-12-20 2006-06-06 Kimberly-Clark Worldwide, Inc. Triggered release from proteinoid microspheres
US20050148497A1 (en) * 2002-02-20 2005-07-07 Khan Mohammed A. Method for administering glp-1 molecules
WO2003080149A2 (en) 2002-03-20 2003-10-02 Mannkind Corporation Inhalation apparatus
US7445796B2 (en) * 2002-08-19 2008-11-04 L. Perrigo Company Pharmaceutically active particles of a monomodal particle size distribution and method
DK1571970T3 (en) * 2002-10-02 2011-11-28 Dmi Biosciences Inc Diagnosis and monitoring of diseases
ES2575563T3 (en) * 2003-05-15 2016-06-29 Ampio Pharmaceuticals, Inc. Treatment of diseases mediated by T lymphocytes
JP3895307B2 (en) * 2003-06-12 2007-03-22 ローム株式会社 Quantitative method and quantitative chip for target substance
US20060286129A1 (en) * 2003-12-19 2006-12-21 Emisphere Technologies, Inc. Oral GLP-1 formulations
JP2005209106A (en) * 2004-01-26 2005-08-04 Nec Corp Portable communication terminal, received e-mail management method, program and recording medium
KR20070008720A (en) * 2004-05-06 2007-01-17 에미스페어 테크놀로지스, 인코포레이티드 Solid dosage form of wetted heparin
KR101299707B1 (en) 2004-05-14 2013-08-28 에미스페어 테크놀로지스, 인코포레이티드 Compounds and compositions for delivering active agents
MXPA06013295A (en) 2004-05-19 2007-02-22 Emisphere Tech Inc Acyclovir formulations.
JP5078014B2 (en) 2004-08-20 2012-11-21 マンカインド コーポレイション Catalytic reaction of diketopiperazine synthesis.
MX2007002189A (en) 2004-08-23 2008-01-11 Mannkind Corp Diketopiperazine salts, diketomorpholine salts or diketodioxane salts for drug delivery.
US9345725B2 (en) 2004-12-29 2016-05-24 Emisphere Technologies, Inc. Pharmaceutical formulations of gallium salts
US7803404B2 (en) 2005-09-14 2010-09-28 Mannkind Corporation Method of drug formulation based on increasing the affinity of active agents for crystalline microparticle surfaces
IN2015DN00888A (en) 2006-02-22 2015-07-10 Mannkind Corp
WO2007121318A2 (en) 2006-04-12 2007-10-25 Emisphere Technologies, Inc. Formulations for delivering insulin
WO2007133944A2 (en) 2006-05-09 2007-11-22 Emisphere Technologies, Inc. Topical administration of acyclovir
WO2008003050A2 (en) 2006-06-28 2008-01-03 Emisphere Technologies, Inc. Gallium nitrate formulations
US8895777B2 (en) * 2006-08-31 2014-11-25 Emisphere Technologies Inc Compounds and compositions for delivering active agents
WO2009146320A1 (en) 2008-05-27 2009-12-03 Dmi Life Sciences, Inc. Therapeutic methods and compounds
US8485180B2 (en) 2008-06-13 2013-07-16 Mannkind Corporation Dry powder drug delivery system
KR101933816B1 (en) 2008-06-13 2019-03-29 맨카인드 코포레이션 A dry powder inhaler and system for drug delivery
WO2009155581A1 (en) 2008-06-20 2009-12-23 Mannkind Corporation An interactive apparatus and method for real-time profiling of inhalation efforts
TWI494123B (en) 2008-08-11 2015-08-01 Mannkind Corp Use of ultrarapid acting insulin
WO2010065362A1 (en) * 2008-11-25 2010-06-10 Innovative Technologies, L.L.C. Improvements in polypeptide synthesis for drug delivery
US8314106B2 (en) 2008-12-29 2012-11-20 Mannkind Corporation Substituted diketopiperazine analogs for use as drug delivery agents
PL2405963T3 (en) 2009-03-11 2014-04-30 Mannkind Corp Apparatus, system and method for measuring resistance of an inhaler
BRPI1013154B1 (en) 2009-06-12 2020-04-07 Mannkind Corp MICROPARTICLES OF DICETOPIPERAZINE WITH SPECIFIC SURFACE AREAS DEFINED, DRY POWDER UNDERSTANDING THE REFERRED MICROPARTICLES, METHOD FOR FORMATION OF THE REFERENCESMICROPARTICLES AND THE FORMATION OF MICROPARTYSTEMS
JP5985985B2 (en) 2009-08-03 2016-09-06 エミスフィアー テクノロジーズ インコーポレイテッドEmisphere Technologies,Inc. Rapid-acting naproxen composition with reduced gastrointestinal effects
CA2778698A1 (en) 2009-11-03 2011-05-12 Mannkind Corporation An apparatus and method for simulating inhalation efforts
MX359281B (en) 2010-06-21 2018-09-21 Mannkind Corp Dry powder drug delivery system and methods.
US8507496B2 (en) 2010-09-07 2013-08-13 Dmi Acquisition Corp. Treatment of diseases
MX353285B (en) 2011-04-01 2018-01-05 Mannkind Corp Blister package for pharmaceutical cartridges.
WO2012174472A1 (en) 2011-06-17 2012-12-20 Mannkind Corporation High capacity diketopiperazine microparticles
PL2766029T3 (en) 2011-10-10 2020-08-24 Ampio Pharmaceuticals, Inc. Treatment of degenerative joint disease
KR20140075772A (en) 2011-10-10 2014-06-19 앰피오 파마슈티컬스 인코퍼레이티드 Implantable medical devices with increased immune tolerance, and methods for making and implanting
IN2014DN03093A (en) 2011-10-24 2015-05-15 Mannkind Corp
SG10201605205VA (en) 2011-10-28 2016-08-30 Ampio Pharmaceuticals Inc Treatment of rhinitis
JP5947527B2 (en) * 2011-11-28 2016-07-06 互応化学工業株式会社 Method for producing cosmetic composition
SG10201605800UA (en) 2012-07-12 2016-09-29 Mannkind Corp Dry powder drug delivery system and methods
SG10202011046RA (en) 2012-09-21 2020-12-30 Intensity Therapeutics Inc Method of treating cancer
WO2014066856A1 (en) 2012-10-26 2014-05-01 Mannkind Corporation Inhalable influenza vaccine compositions and methods
KR20150132508A (en) 2013-03-15 2015-11-25 앰피오 파마슈티컬스 인코퍼레이티드 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
KR102499439B1 (en) 2013-03-15 2023-02-13 맨카인드 코포레이션 Microcrystalline diketopiperazine compositions and methods
CA2918369C (en) 2013-07-18 2021-06-29 Mannkind Corporation Heat-stable dry powder pharmaceutical compositions and methods
WO2015021064A1 (en) 2013-08-05 2015-02-12 Mannkind Corporation Insufflation apparatus and methods
US10307464B2 (en) 2014-03-28 2019-06-04 Mannkind Corporation Use of ultrarapid acting insulin
RU2736513C2 (en) 2014-08-18 2020-11-17 Ампио Фармасьютикалз, Инк. Treating pathological conditions of joints
US10561806B2 (en) 2014-10-02 2020-02-18 Mannkind Corporation Mouthpiece cover for an inhaler
US11389512B2 (en) 2015-06-22 2022-07-19 Ampio Pharmaceuticals, Inc. Use of low molecular weight fractions of human serum albumin in treating diseases

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2133926A1 (en) * 1971-04-21 1972-12-01 Yeda Research Develop Co
DE2424169A1 (en) * 1973-05-17 1974-12-05 Little Inc A IMPLANT BODY AND METHOD OF ITS MANUFACTURING
US4348384A (en) * 1980-10-17 1982-09-07 Dainippon Pharmaceutical Co., Ltd. Pharmaceutical composition for oral administration containing coagulation factor VIII or IX
EP0105804A2 (en) * 1982-09-30 1984-04-18 The University Of Rochester Human monoclonal antibodies against bacterial toxins
DE3612102A1 (en) * 1985-04-10 1986-10-16 Československá akademie věd, Prag/Praha SOLUBLE AND BIODEGRADABLE MIXED POLYMERS FOR THE BINDING OF BIOLOGICALLY ACTIVE SUBSTANCES AND METHOD FOR THE PRODUCTION THEREOF
WO1987004076A1 (en) * 1985-12-30 1987-07-16 The Texas A & M University System Low dosage of interferon to enhance vaccine efficiency
WO1988001213A1 (en) * 1986-08-18 1988-02-25 Clinical Technologies Associates, Inc. Delivery systems for pharmacological agents
EP0366277A2 (en) * 1988-09-29 1990-05-02 Patralan Limited Pharmaceutical formulations
US4976968A (en) * 1989-02-24 1990-12-11 Clinical Technologies Associates, Inc. Anhydrous delivery systems for pharmacological agents
US4983402A (en) * 1989-02-24 1991-01-08 Clinical Technologies Associates, Inc. Orally administerable ANF
EP0448057A1 (en) * 1990-03-19 1991-09-25 Bristol-Myers Squibb Company Monoclonal antibody reactive with a novel HLA determinant on MHC class I molecules and method for activating lymphocytes
EP0452161A1 (en) * 1990-02-12 1991-10-16 Elf Sanofi Cosmetic composition containing amino acids copolymers, useful as hydrating agent
US5069936A (en) * 1987-06-25 1991-12-03 Yen Richard C K Manufacturing protein microspheres
EP0459795A1 (en) * 1990-06-01 1991-12-04 Kirin-Amgen, Inc. Oral dosage form of biologically active proteins
EP0467389A2 (en) * 1990-07-19 1992-01-22 University Of Kentucky Research Foundation Drug delivery system involving interaction between protein or polypeptide and hydrophobic biodegradable polymer

Family Cites Families (175)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2671451A (en) * 1952-06-16 1954-03-09 Stephen J Bolger Remedial pill
BE530009A (en) * 1953-06-30
US2828206A (en) 1954-02-24 1958-03-25 Roseuberg Adolf Stabilized fat-soluble vitamins and methods of making same
US2868740A (en) * 1954-03-25 1959-01-13 Swift & Co Method of copolymerizing acrylic or methacrylic acid with proteinaceous material and product obtained
US2862918A (en) * 1956-03-12 1958-12-02 Glidden Co Acylated, isolated, partially-hydrolyzed, soya protein and process
NL108169C (en) * 1957-01-30
US3057344A (en) * 1957-05-21 1962-10-09 Abella Carlos Alberto Capsule for the study of the digestive tract and method of using the same
US3016308A (en) * 1957-08-06 1962-01-09 Moore Business Forms Inc Recording paper coated with microscopic capsules of coloring material, capsules and method of making
US3052655A (en) * 1958-08-01 1962-09-04 Sidney W Fox Thermal polymerization of amino acid mixtures containing aspartic acid or a thermal precursor of aspartic acid
US3076790A (en) * 1958-08-01 1963-02-05 Sidney W Fox Method of making copolymers of amino acids containing glutamic acid
GB929401A (en) 1958-12-22 1963-06-19 Upjohn Co Encapsulated emulsions and processes for their preparation
FR1468601A (en) 1958-12-22 1967-02-10 Ncr Co Process for forming protective coatings for solid and liquid particles
FR1351358A (en) 1958-12-22 1964-02-07 Ncr Co Process for forming impermeable coatings for particulate matter by liquid phase separation
NL129921C (en) * 1958-12-31
US3170802A (en) * 1960-12-14 1965-02-23 Zh Noda Sangyo Kagaku Kenkyush Method for treatment of soybean proteins
GB1075952A (en) 1962-12-31 1967-07-19 Gelatine And Glue Res Ass Microscopic capsules and methods of making them
US3748277A (en) * 1965-10-14 1973-07-24 Ncr Co Process of forming minute capsules
US3474777A (en) * 1966-02-10 1969-10-28 Amp Inc Method of administering therapeutic agents
US3576758A (en) * 1966-10-17 1971-04-27 Ncr Co Treatment of polypeptide-containing hydrophilic polymeric capsule wall material with uranium and vanadium compounds
JPS4813070B1 (en) * 1967-04-20 1973-04-25
FR7981M (en) 1967-10-21 1970-06-08
US3491093A (en) * 1967-11-29 1970-01-20 Endo Lab Derivatives of 5 aminomethyl-4,5,6,7-tetrahydro-4-oxoindoles
US3565559A (en) * 1968-03-11 1971-02-23 Sumitomo Chemical Co Process for making microcapsules
US3574832A (en) * 1968-05-29 1971-04-13 American Cyanamid Co Therapeutic heparin-surfactant compositions
GB1236885A (en) 1968-09-28 1971-06-23 Fuji Photo Film Co Ltd Method of making multi-wall capsules
US3567650A (en) * 1969-02-14 1971-03-02 Ncr Co Method of making microscopic capsules
US3937668A (en) * 1970-07-15 1976-02-10 Ilse Zolle Method for incorporating substances into protein microspheres
US3725113A (en) * 1970-12-17 1973-04-03 Research Corp Blood compatible microencapsulated detoxicants and method for making
US3822348A (en) * 1970-12-28 1974-07-02 Toyo Jozo Kk Hormone-like substance having serum calcium reducing property
US3962416A (en) * 1971-01-25 1976-06-08 Sol Katzen Preserved nutrients and products
US3794561A (en) * 1971-09-30 1974-02-26 Sasaki T Biologically active peptide and method of preparing the same
US3933873A (en) * 1971-12-08 1976-01-20 Texaco Inc. Preparation of omega-aminoalkanoic acids
US3816404A (en) * 1971-12-08 1974-06-11 Texaco Inc Preparation of caprolactam
US3795739A (en) * 1972-02-14 1974-03-05 Hoffmann La Roche Treatment of parkinson disease
JPS5210427B2 (en) * 1972-07-19 1977-03-24
US4351337A (en) * 1973-05-17 1982-09-28 Arthur D. Little, Inc. Biodegradable, implantable drug delivery device, and process for preparing and using the same
US4450150A (en) * 1973-05-17 1984-05-22 Arthur D. Little, Inc. Biodegradable, implantable drug delivery depots, and method for preparing and using the same
DE2343037A1 (en) 1973-08-25 1975-03-06 Hoechst Ag MEDICINAL PRODUCTS WITH ANTIDEPRESSIVE EFFECT
US3939253A (en) * 1973-11-02 1976-02-17 Interx Research Corporation Novel, transient pro-drug forms of l-dopa useful in the treatment of parkinson's disease
GB1459488A (en) * 1974-03-19 1976-12-22 Wyeth John & Brother Ltd Piperazinedione derivatives
US4061466A (en) * 1974-10-16 1977-12-06 Ingvar Gosta Holger Sjoholm Biologically active composition and the use thereof
US4183849A (en) * 1975-01-15 1980-01-15 Nordisk Insulinlaboratorium Therapeutic insulin preparation and a process for the production of a stable insulin preparation with protracted effect
US4048268A (en) * 1975-02-19 1977-09-13 Eli Lilly And Company Stabilization method
US4035507A (en) * 1975-04-17 1977-07-12 Interx Research Corporation Novel, transient pro-drug forms of L-DOPA to treat Parkinson's disease
DE2517229A1 (en) 1975-04-18 1976-10-28 Boehringer Mannheim Gmbh PHENYLALKYLCARBONIC ACID DERIVATIVES AND PROCESS FOR THEIR PRODUCTION
CA1077842A (en) 1975-10-09 1980-05-20 Minnesota Mining And Manufacturing Company Albumin medicament carrier system
US4405598A (en) * 1976-01-30 1983-09-20 Fisons, Limited Composition for treating asthma
US4117801A (en) * 1976-06-10 1978-10-03 Eastman Kodak Company Apparatus for spray coating discrete particles
US4061507A (en) * 1976-06-28 1977-12-06 Richmond Industries, Inc. Wrench and method of making the same
FR2374910A1 (en) 1976-10-23 1978-07-21 Choay Sa PREPARATION BASED ON HEPARIN, INCLUDING LIPOSOMES, PROCESS FOR OBTAINING IT AND MEDICINAL PRODUCTS CONTAINING SUCH PREPARATIONS
US4357259A (en) 1977-08-01 1982-11-02 Northwestern University Method of incorporating water-soluble heat-sensitive therapeutic agents in albumin microspheres
US4217370A (en) * 1977-08-25 1980-08-12 Blue Wing Corporation Lipid-containing feed supplements and foodstuffs
US4199561A (en) * 1979-02-26 1980-04-22 The Dow Chemical Company Coated nutrients and medicaments for veterinary use
US4352883A (en) * 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US4345588A (en) * 1979-04-23 1982-08-24 Northwestern University Method of delivering a therapeutic agent to a target capillary bed
US4239635A (en) * 1979-06-11 1980-12-16 Cincinnati Milacron Inc. Novel diamide and lubricants containing same
US4272506A (en) * 1979-08-31 1981-06-09 Syva Company Purification of reagents by disulfide immobilization
HU181009B (en) * 1980-01-18 1983-05-30 Richter Gedeon Vegyeszet Process for preparing angiotensin-ii analogues with antagonictic activity containing in position 1 sarcosyl,hydroxyacetyl or l-alpha-aminoxy-propionyl group and in positiona 8 esteric group
NZ196349A (en) 1980-03-07 1984-08-24 Interx Research Corp Enhancement of absorption rate of orally administered polar bioactive agents
IT1148784B (en) * 1980-04-09 1986-12-03 Eurand Spa PROCEDURE FOR THE PREPARATION OF MICRO CAPSULES IN A LIQUID VEHICLE
DE3016170A1 (en) * 1980-04-26 1981-10-29 Bayer Ag, 5090 Leverkusen MICROCAPSULES WITH A DEFINED OPENING TEMPERATURE, METHOD FOR THE PRODUCTION AND USE THEREOF
CA1155853A (en) 1980-06-06 1983-10-25 Joseph A. Martin Imidazole derivatives and preparation thereof
US4289759A (en) * 1980-06-23 1981-09-15 Ortho Pharmaceutical Corporation Immunoregulatory diketopiperazine compounds
US4442090A (en) * 1980-11-09 1984-04-10 Kyoto Yakuhin Kogyo Kabushiki Kaisha Absorption-promoting compounds, compositions thereof with pharmaceuticals and/or bases for rectal administration and method of use
US4900730A (en) * 1981-01-14 1990-02-13 Toyo Jozo Co., Ltd. Preparation which promotes the absorption of peptides
GB2092136B (en) * 1981-01-17 1985-06-05 Mitsui Toatsu Chemicals Production of n-substituted amide compounds
US4483807A (en) 1981-01-27 1984-11-20 Japan Atomic Energy Research Institute Process for producing a slow release composite
FR2509175B1 (en) 1981-03-06 1987-01-16 Toyo Jozo Kk THERAPEUTIC PREPARATION HAVING EXCELLENT ABSORPTION PROPERTIES
JPS58140026A (en) * 1982-01-14 1983-08-19 Toyo Jozo Co Ltd Pharmaceutical having improved absorbability
NZ201010A (en) 1981-06-19 1986-02-21 Ciba Geigy Ag The treatment of inflammation diseases using desferrioxamine
US4446138A (en) * 1982-02-10 1984-05-01 Pack Howard M Method and composition for reducing weight
CA1241646A (en) * 1982-02-22 1988-09-06 Adolfo J. De Bold Atrial natriuretic factor
US4457907A (en) 1982-08-05 1984-07-03 Clear Lake Development Group Composition and method for protecting a therapeutic drug
US4518433A (en) * 1982-11-08 1985-05-21 Fmc Corporation Enteric coating for pharmaceutical dosage forms
US4393192A (en) * 1982-12-21 1983-07-12 The Standard Oil Company Crystalline copolymers prepared from N,N'-terephthaloyldi-beta-alanine and a glycol
US4473620A (en) * 1982-12-23 1984-09-25 Eastman Kodak Company Encapsulated butylated hydroxyanisole
US4886663A (en) * 1983-01-03 1989-12-12 Scripps Clinic And Research Foundation Synthetic heat-stable enterotoxin polypeptide of Escherichia coli and multimers thereof
JPS59163313A (en) * 1983-03-09 1984-09-14 Teijin Ltd Peptide hormone composition for nasal administration
CA1196862A (en) * 1983-06-01 1985-11-19 Anthony M.F. Sun Microencapsulation of living tissue and cells
CA1196863A (en) * 1983-06-08 1985-11-19 Mattheus F.A. Goosen Slow release injectable insulin composition
US4462839A (en) * 1983-06-16 1984-07-31 Fmc Corporation Enteric coating for pharmaceutical dosage forms
US4608278A (en) * 1983-06-22 1986-08-26 The Ohio State University Research Foundation Small particule formation and encapsulation
ATE29392T1 (en) 1983-06-22 1987-09-15 Univ Ohio State Res Found PREPARATION OF A FINE PARTICULATED MATERIAL AND ENCAPSULATION.
US4692433A (en) 1983-10-12 1987-09-08 The Regents Of The University Of California Method and composition for regulating serum calcium levels of mammals
JPS60125245A (en) * 1983-12-12 1985-07-04 Nitto Electric Ind Co Ltd Preparation of microcapsule containing liquid active substance
US4671954A (en) * 1983-12-13 1987-06-09 University Of Florida Microspheres for incorporation of therapeutic substances and methods of preparation thereof
US4590265A (en) * 1984-02-17 1986-05-20 Eastman Kodak Company Carboxylated cellulose ester and manufacture thereof
JPS60176549A (en) * 1984-02-22 1985-09-10 Nisshin Oil Mills Ltd:The Preparation of protein hydrolyzate
US4703042A (en) * 1984-05-21 1987-10-27 Bodor Nicholas S Orally active heparin salts containing multivalent cationic units
FR2565102B1 (en) * 1984-06-05 1987-03-20 Paris Sud Universite BIODEGRADABLE MICROCAPSULES BASED ON SERUMALBUMIN, THEIR PREPARATION AND THEIR APPLICATION TO THE IN SITU RELEASE OF MEDICUMENTS
US4757066A (en) * 1984-10-15 1988-07-12 Sankyo Company Limited Composition containing a penem or carbapenem antibiotic and the use of the same
IT1177384B (en) * 1984-12-12 1987-08-26 Boeehringer Biochemia Robin Sp DIETARY GRANULAR PRODUCTS BASED ON AMINO ACIDS AND PROCEDURE FOR THEIR PREPARATION
US4708952A (en) * 1985-02-06 1987-11-24 Aida Salatinjants Method of treatment of the infectious and viral diseases by one time interference
US4908233A (en) 1985-05-08 1990-03-13 Lion Corporation Production of microcapsules by simple coacervation
US4757024A (en) * 1985-05-31 1988-07-12 Biostar Medical Products, Inc. Immune complex detection method and article using immunologically non-specific immunoglobulins
US4897444A (en) * 1985-05-31 1990-01-30 The Research Foundation Of The State University Of New York Immobilized fluorogenic substrates for enzymes; and processes for their preparation
US4683092A (en) 1985-07-03 1987-07-28 Damon Biotech, Inc. Capsule loading technique
US4789734A (en) * 1985-08-06 1988-12-06 La Jolla Cancer Research Foundation Vitronectin specific cell receptor derived from mammalian mesenchymal tissue
IT1214629B (en) * 1985-08-29 1990-01-18 Formenti Farmaceutici Spa MICRO-ENCAPSULATION PROCEDURE OF A MEDICATION, MEDICATION SO PREPARED, AND PHARMACEUTICAL COMPOSITIONS THAT INCLUDE IT
DE3682257D1 (en) * 1985-11-22 1991-12-05 Takeda Chemical Industries Ltd LIPOSOME COMPOSITION.
LU86258A1 (en) 1986-01-21 1987-09-03 Rech Dermatologiques C I R D S BENZAMIDO AROMATIC COMPOUNDS, PROCESS FOR THEIR PREPARATION AND THEIR USE IN HUMAN OR VETERINARY MEDICINE AND IN COSMETICS
IT1188550B (en) * 1986-02-07 1988-01-14 Sclavo Spa SYNTHETIC PEPTIDE WITH INTERLEUKINA 1 HUMAN ACTIVITY
US4919939A (en) * 1986-04-29 1990-04-24 Pharmetrix Corporation Periodontal disease treatment system
US4692284A (en) * 1986-04-30 1987-09-08 Damon Biotech, Inc. Method and apparatus for forming droplets and microcapsules
NZ221411A (en) 1986-08-11 1989-10-27 Innovata Biomed Ltd Pharmaceutical compositions containing microcapsules and a surfactant
US4837381A (en) * 1986-08-11 1989-06-06 American Cyanamid Company Compositions for parenteral administration and their use
CH668257A5 (en) * 1986-09-23 1988-12-15 Moeller Willi Fa DICARBONIC ACID DIAMOND, THESE CONTAINING ION SELECTIVE MEMBRANES AND TEST DEVICES, AND LITHIUM COMPLEXES OF DICARBONIC ACID DIAMOND.
DE3700128A1 (en) * 1987-01-03 1988-07-14 Hoechst Ag BIODEGRADABLE POLY- (HYDROXYALKYL) - AMINODICARBONIC ACID DERIVATIVES, METHOD FOR THE PRODUCTION AND USE THEREOF FOR DEPOT PREPARATIONS WITH CONTROLLED ACTIVE SUBSTANCE DELIVERY
US5077278A (en) * 1987-01-23 1991-12-31 Pfizer Inc. Non-natural demethylavermectins compositions and method of use
MX12394A (en) 1987-07-23 1993-12-01 Ciba Geigy Ag PROCEDURE FOR OBTAINING POLYETHYLENE GLYCOL CARBAMATES.
JPH0725725B2 (en) 1987-07-23 1995-03-22 保土谷化学工業株式会社 Benzamide derivative
US4895725A (en) * 1987-08-24 1990-01-23 Clinical Technologies Associates, Inc. Microencapsulation of fish oil
US5067961A (en) * 1988-02-18 1991-11-26 Autogenesis Technologies, Inc. Non-biodegradable two phase corneal implant and method for preparing same
JP2670680B2 (en) * 1988-02-24 1997-10-29 株式会社ビーエムジー Polylactic acid microspheres containing physiologically active substance and method for producing the same
GB8811409D0 (en) 1988-05-13 1988-06-15 Unilever Plc Cosmetic composition
GB8811408D0 (en) 1988-05-13 1988-06-15 Unilever Plc Cosmetic composition
US5055300A (en) * 1988-06-17 1991-10-08 Basic Bio Systems, Inc. Time release protein
FR2636238B1 (en) * 1988-09-14 1994-01-21 Morelle Jean NEW ANTISUDORAL COMPOSITIONS
GB8823731D0 (en) 1988-10-10 1988-11-16 Smith Kline French Lab Biologically active compounds
US5039481A (en) * 1988-12-16 1991-08-13 Clean Air, Inc. Aliphatic polycarboxylic acids as air purification compositions
CA2012306A1 (en) 1989-03-28 1990-09-28 Werner Neidhart Amino acid derivatives
US5122367A (en) * 1989-03-31 1992-06-16 Massachusetts Institute Of Technology Polyanhydride bioerodible controlled release implants for administration of stabilized growth hormone
US4963364A (en) * 1989-04-10 1990-10-16 Fox Sidney W Microencapsulated antitumor agent
US5019400A (en) * 1989-05-01 1991-05-28 Enzytech, Inc. Very low temperature casting of controlled release microspheres
US5100918A (en) * 1989-05-25 1992-03-31 Sterling Drug, Inc. Prevention or treatment of sunburn using the S(+) isomer of ibuprofen
US4996292A (en) * 1989-06-30 1991-02-26 Fox Sidney W Self-sealing artificial skin comprising copoly-alpha-amino acid
JP2911496B2 (en) 1989-09-11 1999-06-23 帝國製薬株式会社 Highly absorbable vaginal agent containing bioactive polypeptide
US5271961A (en) 1989-11-06 1993-12-21 Alkermes Controlled Therapeutics, Inc. Method for producing protein microspheres
US5216124A (en) 1989-12-15 1993-06-01 G. D. Searle & Co. Substituted cyclic tetrapeptides
US5389377A (en) 1989-12-22 1995-02-14 Molecular Bioquest, Inc. Solid care therapeutic compositions and methods for making same
US5126147A (en) * 1990-02-08 1992-06-30 Biosearch, Inc. Sustained release dosage form
GB9007052D0 (en) 1990-03-29 1990-05-30 Skua Investments Ltd Pharmaceutical formulations
US5451410A (en) 1993-04-22 1995-09-19 Emisphere Technologies, Inc. Modified amino acids for encapsulating active agents
US5714167A (en) 1992-06-15 1998-02-03 Emisphere Technologies, Inc. Active agent transport systems
US5578323A (en) 1992-06-15 1996-11-26 Emisphere Technologies, Inc. Proteinoid carriers and methods for preparation and use thereof
US5693338A (en) 1994-09-29 1997-12-02 Emisphere Technologies, Inc. Diketopiperazine-based delivery systems
US5629020A (en) 1994-04-22 1997-05-13 Emisphere Technologies, Inc. Modified amino acids for drug delivery
US5443841A (en) 1992-06-15 1995-08-22 Emisphere Technologies, Inc. Proteinoid microspheres and methods for preparation and use thereof
US5541155A (en) 1994-04-22 1996-07-30 Emisphere Technologies, Inc. Acids and acid salts and their use in delivery systems
US5447728A (en) 1992-06-15 1995-09-05 Emisphere Technologies, Inc. Desferrioxamine oral delivery system
JPH05239021A (en) 1990-09-04 1993-09-17 Microbial Chem Res Found New actinonin derivative
US5418010A (en) 1990-10-05 1995-05-23 Griffith Laboratories Worldwide, Inc. Microencapsulation process
DE4033419A1 (en) 1990-10-20 1992-04-23 Wolman Gmbh Dr POLYMOUS NITROGEN COMPOUNDS AND METAL FIXING SAEURS CONTAINING WOOD PROTECTION AGENTS
US5271934A (en) 1990-10-22 1993-12-21 Revlon Consumer Products Corporation Encapsulated antiperspirant salts and deodorant/antiperspirants
JP3179538B2 (en) 1990-12-11 2001-06-25 ノバルティス アクチエンゲゼルシャフト Aqueous solution of stable human calcitonin
US5137892A (en) * 1990-12-12 1992-08-11 Abbott Laboratories Quinoline, naphthyridine and pyridobenzoxazine derivatives
US5244653A (en) 1991-05-01 1993-09-14 Isp Chemicals Inc. Glycine anhydride dimethylol as a biocide and preservative
CA2070061C (en) 1991-06-07 2004-02-10 Shigeyuki Takama Physiologically active polypeptide-containing pharmaceutical composition
US5250236A (en) 1991-08-05 1993-10-05 Gasco Maria R Method for producing solid lipid microspheres having a narrow size distribution
ZA93929B (en) 1992-02-18 1993-09-10 Akzo Nv A process for the preparation of biologically active materialcontaining polymeric microcapsules.
US5352461A (en) 1992-03-11 1994-10-04 Pharmaceutical Discovery Corporation Self assembling diketopiperazine drug delivery system
US5310535A (en) 1992-04-24 1994-05-10 The Dow Chemical Company Carboxamide modified polyamine chelators and radioactive complexes thereof for conjugation to antibodies
US5792451A (en) 1994-03-02 1998-08-11 Emisphere Technologies, Inc. Oral drug delivery compositions and methods
US5811127A (en) 1992-06-15 1998-09-22 Emisphere Technologies, Inc. Desferrioxamine oral delivery system
HU211995B (en) 1992-06-30 1996-01-29 Gyogyszerkutato Intezet Process to prepare novel benzoyl amino acid derivs. and pharmaceutical compns. contg.them
US5401516A (en) 1992-12-21 1995-03-28 Emisphere Technologies, Inc. Modified hydrolyzed vegetable protein microspheres and methods for preparation and use thereof
AU6096094A (en) 1993-01-27 1994-08-15 Georgetown University Method and composition employing (2r,4s) itraconazole
US5439686A (en) 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
DK0616799T3 (en) 1993-03-24 2000-09-18 Collaborative Lab Inc Cosmetic application system for salicylic acid and process for their preparation
AU6819294A (en) 1993-04-22 1994-11-08 Emisphere Technologies, Inc. Oral drug delivery compositions and methods
US5643957A (en) 1993-04-22 1997-07-01 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5709861A (en) 1993-04-22 1998-01-20 Emisphere Technologies, Inc. Compositions for the delivery of antigens
ATE171715T1 (en) 1993-04-23 1998-10-15 Rhodia Chimie Sa POLYANHYDROASPARAGIC ACID AND ITS BIODEGRADABLE HYDROLYSIS PRODUCTS
US5650386A (en) 1995-03-31 1997-07-22 Emisphere Technologies, Inc. Compositions for oral delivery of active agents
US5866536A (en) 1995-03-31 1999-02-02 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5820881A (en) 1995-04-28 1998-10-13 Emisphere Technologies, Inc. Microspheres of diamide-dicarboxylic acids
US5667806A (en) 1995-06-07 1997-09-16 Emisphere Technologies, Inc. Spray drying method and apparatus
US5824345A (en) 1995-06-07 1998-10-20 Emisphere Technologies, Inc. Fragrances and flavorants
US5750147A (en) 1995-06-07 1998-05-12 Emisphere Technologies, Inc. Method of solubilizing and encapsulating itraconazole
US5773647A (en) 1997-02-07 1998-06-30 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5879681A (en) 1997-02-07 1999-03-09 Emisphere Technolgies Inc. Compounds and compositions for delivering active agents
US5876710A (en) 1997-02-07 1999-03-02 Emisphere Technologies Inc. Compounds and compositions for delivering active agents
US5804688A (en) 1997-02-07 1998-09-08 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5776888A (en) 1997-02-07 1998-07-07 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5863944A (en) 1997-04-30 1999-01-26 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2133926A1 (en) * 1971-04-21 1972-12-01 Yeda Research Develop Co
DE2424169A1 (en) * 1973-05-17 1974-12-05 Little Inc A IMPLANT BODY AND METHOD OF ITS MANUFACTURING
US4348384A (en) * 1980-10-17 1982-09-07 Dainippon Pharmaceutical Co., Ltd. Pharmaceutical composition for oral administration containing coagulation factor VIII or IX
EP0105804A2 (en) * 1982-09-30 1984-04-18 The University Of Rochester Human monoclonal antibodies against bacterial toxins
DE3612102A1 (en) * 1985-04-10 1986-10-16 Československá akademie věd, Prag/Praha SOLUBLE AND BIODEGRADABLE MIXED POLYMERS FOR THE BINDING OF BIOLOGICALLY ACTIVE SUBSTANCES AND METHOD FOR THE PRODUCTION THEREOF
WO1987004076A1 (en) * 1985-12-30 1987-07-16 The Texas A & M University System Low dosage of interferon to enhance vaccine efficiency
WO1988001213A1 (en) * 1986-08-18 1988-02-25 Clinical Technologies Associates, Inc. Delivery systems for pharmacological agents
US5069936A (en) * 1987-06-25 1991-12-03 Yen Richard C K Manufacturing protein microspheres
EP0366277A2 (en) * 1988-09-29 1990-05-02 Patralan Limited Pharmaceutical formulations
US4976968A (en) * 1989-02-24 1990-12-11 Clinical Technologies Associates, Inc. Anhydrous delivery systems for pharmacological agents
US4983402A (en) * 1989-02-24 1991-01-08 Clinical Technologies Associates, Inc. Orally administerable ANF
EP0452161A1 (en) * 1990-02-12 1991-10-16 Elf Sanofi Cosmetic composition containing amino acids copolymers, useful as hydrating agent
EP0448057A1 (en) * 1990-03-19 1991-09-25 Bristol-Myers Squibb Company Monoclonal antibody reactive with a novel HLA determinant on MHC class I molecules and method for activating lymphocytes
EP0459795A1 (en) * 1990-06-01 1991-12-04 Kirin-Amgen, Inc. Oral dosage form of biologically active proteins
EP0467389A2 (en) * 1990-07-19 1992-01-22 University Of Kentucky Research Foundation Drug delivery system involving interaction between protein or polypeptide and hydrophobic biodegradable polymer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BIOCHEMISTRY vol. 3, no. 11 , November 1964 , EASTON, PA US pages 1665 - 1674 G.D. FASMAN ET AL. 'Optical Rotatory Dispersion Studies of Poly-L-tyrosine and Copolymers of L-Glutamic Acid and L-Tyrosine. Significance of the Tyrosyl Cotton Effects with Respect to Protein Conformation' *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5869466A (en) * 1993-05-24 1999-02-09 Biotech Australia Pty Limited Vitamin B12 mediated oral delivery systems for GCSF
US6262253B1 (en) * 1993-05-24 2001-07-17 Biotech Australia Pty Limited Vitamin B12 conjugates with gcsf, analogues thereof and pharmaceutical compositions
US5548064A (en) * 1993-05-24 1996-08-20 Biotech Australia Pty Limited Vitamin B12 conjugates with EPO, analogues thereof and pharmaceutical compositions
EP0706375A4 (en) * 1993-06-14 1996-11-06 Emisphere Tech Inc Proteinoid carriers
EP0706375A1 (en) * 1993-06-14 1996-04-17 Emisphere Technologies, Inc. Proteinoid carriers
US5935601A (en) * 1994-04-22 1999-08-10 Emisphere Technologies, Inc. Modified amino acids for drug delivery
EP0706798A1 (en) * 1994-07-22 1996-04-17 Sanwa Kagaku Kenkyusho Co., Ltd. Pharmaceutical composition containing biologically active peptide or protein
EP0831784A1 (en) * 1995-06-07 1998-04-01 Emisphere Technologies, Inc. Fragrances and flavorants
EP0831784A4 (en) * 1995-06-07 2000-07-19 Emisphere Tech Inc Fragrances and flavorants
WO1999064495A1 (en) * 1998-06-05 1999-12-16 Ústav Makromolekulární Chemie Akademie Věd České Republiky FUNCTIONALISED POLYMERS OF α-AMINO ACIDS AND THE METHOD OF PREPARATION THEREOF
US6590061B1 (en) * 1998-06-05 2003-07-08 Institute Of Macromolecular Chemistry Of The Academy Of Sciences Of The Czech Republic Functionalized polymers of α-amino acids and the method of preparation thereof
US6652875B1 (en) 1998-07-29 2003-11-25 Pacific Biolink Pty. Limited Casein formulations for the delivery of bioactive constituents
US7282216B2 (en) 2001-11-12 2007-10-16 Alkermes Controlled Therapeutics, Inc. Biocompatible polymer blends and uses thereof

Also Published As

Publication number Publication date
EP0642532A1 (en) 1995-03-15
US6413550B1 (en) 2002-07-02
KR950701939A (en) 1995-05-17
CA2138146A1 (en) 1993-12-23
WO1993025583A3 (en) 1994-08-04
HU9403589D0 (en) 1995-02-28
US5578323A (en) 1996-11-26
CZ315494A3 (en) 1996-01-17
FI945912A0 (en) 1994-12-15
FI945912A (en) 1995-02-15
US5840340A (en) 1998-11-24
HUT70211A (en) 1995-09-28
BR9306678A (en) 1998-12-08
AU4635693A (en) 1994-01-04
JPH07508004A (en) 1995-09-07

Similar Documents

Publication Publication Date Title
US6413550B1 (en) Proteinoid carriers and methods for preparation and use thereof
WO1993025583A9 (en) Proteinoid carriers and methods for preparation and use thereof
US5443841A (en) Proteinoid microspheres and methods for preparation and use thereof
CA2174961C (en) Desferrioxamine oral delivery system
US5811127A (en) Desferrioxamine oral delivery system
US5451410A (en) Modified amino acids for encapsulating active agents
US5401516A (en) Modified hydrolyzed vegetable protein microspheres and methods for preparation and use thereof
US5352461A (en) Self assembling diketopiperazine drug delivery system
TW555569B (en) Biodegradable microparticles for the sustained delivery of therapeutic drugs
US6221397B1 (en) Surface cross-linked particles suitable for controlled delivery
JPH08509474A (en) Oral drug transfer composition and method thereof
CN103429267B (en) Branched polymer aggregate of hydrophobic molecule induction and uses thereof
CA2160692A1 (en) Modified hydrolyzed vegetable protein microspheres and methods for preparation and use thereof
WO1994014420A9 (en) Modified hydrolyzed vegetable protein microspheres and methods for preparation and use thereof
AU1186401A (en) Ionically formulated biomolecule microcarriers
AU697044B2 (en) Proteinoid carriers
JPH0386834A (en) New pharmaceutical of peptide or protein for oral medicine
Kwon Thermosensitive biodegradable hydrogels for the delivery of therapeutic agents
AU8995198A (en) Cross-linked particles

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AT AU BB BG BR CA CH CZ DE DK ES FI GB HU JP KP KR KZ LK LU MG MN MW NL NO NZ PL RO RU SD SE SK UA US VN

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGES 57-70,DESCRIPTION,REPLACED BY NEW PAGES 57-71;PAGES 71-76,CLAIMS,RENUMBERED AS PAGES 72-77;PAGES 1/28-28/28,DRAWINGS,REPLACED BY NEW PAGES 1/23-23/23

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AT AU BB BG BR CA CH CZ DE DK ES FI GB HU JP KP KR KZ LK LU MG MN MW NL NO NZ PL RO RU SD SE SK UA US VN

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 254375

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2138146

Country of ref document: CA

Ref document number: PV1994-3154

Country of ref document: CZ

WWE Wipo information: entry into national phase

Ref document number: 1993916542

Country of ref document: EP

Ref document number: 945912

Country of ref document: FI

WWP Wipo information: published in national office

Ref document number: 1993916542

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: PV1994-3154

Country of ref document: CZ

WWR Wipo information: refused in national office

Ref document number: PV1994-3154

Country of ref document: CZ

WWW Wipo information: withdrawn in national office

Ref document number: 1993916542

Country of ref document: EP