US20010039258A1 - Active agent transport systems - Google Patents

Active agent transport systems Download PDF

Info

Publication number
US20010039258A1
US20010039258A1 US09/760,307 US76030701A US2001039258A1 US 20010039258 A1 US20010039258 A1 US 20010039258A1 US 76030701 A US76030701 A US 76030701A US 2001039258 A1 US2001039258 A1 US 2001039258A1
Authority
US
United States
Prior art keywords
perturbant
active agent
biologically active
alkyl
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/760,307
Inventor
Sam Milstein
Leone-Bay Andrea
Donald Sarubbi
Harry Leipold
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Emisphere Technologies Inc
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
Priority claimed from US08/051,019 external-priority patent/US5451410A/en
Priority claimed from US08/076,803 external-priority patent/US5578323A/en
Priority claimed from US08/168,776 external-priority patent/US5447728A/en
Priority claimed from US08/231,622 external-priority patent/US5629020A/en
Priority claimed from PCT/US1994/004560 external-priority patent/WO1994023767A1/en
Priority claimed from US08/231,623 external-priority patent/US5541155A/en
Priority claimed from US08/315,200 external-priority patent/US5693338A/en
Priority claimed from US08/316,404 external-priority patent/US6331318B1/en
Priority claimed from US08/328,932 external-priority patent/US5714167A/en
Priority claimed from US08/763,183 external-priority patent/US6099856A/en
Priority claimed from US08/820,694 external-priority patent/US6344213B1/en
Priority to US09/760,307 priority Critical patent/US20010039258A1/en
Application filed by Emisphere Technologies Inc filed Critical Emisphere Technologies Inc
Publication of US20010039258A1 publication Critical patent/US20010039258A1/en
Assigned to MHR INSTITUTIONAL PARTNERS IIA LP reassignment MHR INSTITUTIONAL PARTNERS IIA LP SECURITY AGREEMENT Assignors: EMISPHERE TECHNOLOGIES, INC.
Abandoned legal-status Critical Current

Links

Images

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/726Glycosaminoglycans, i.e. mucopolysaccharides
    • A61K31/727Heparin; Heparan
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/23Calcitonins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/25Growth hormone-releasing factor [GH-RF] (Somatoliberin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/27Growth hormone [GH] (Somatotropin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/29Parathyroid hormone (parathormone); Parathyroid hormone-related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/541Organic ions forming an ion pair complex with the pharmacologically or therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • A61K9/1617Organic compounds, e.g. phospholipids, fats
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • 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
    • C07C233/46Carboxylic 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 an acyclic carbon atom
    • C07C233/47Carboxylic 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 an acyclic carbon atom having the carbon atom of the carboxamide group bound to a hydrogen atom or to a carbon atom of an acyclic saturated carbon skeleton
    • 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
    • C07C233/46Carboxylic 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 an acyclic carbon atom
    • C07C233/48Carboxylic 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 an acyclic carbon atom having the carbon atom of the carboxamide group bound to an acyclic carbon atom of a saturated carbon skeleton containing rings
    • 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
    • C07C233/46Carboxylic 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 an acyclic carbon atom
    • C07C233/51Carboxylic 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 an acyclic carbon atom having the carbon atom of the carboxamide group bound to an acyclic carbon atom of a carbon skeleton containing six-membered aromatic rings
    • 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
    • C07C233/52Carboxylic 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 ring other than a six-membered aromatic ring
    • 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
    • C07C233/83Carboxylic 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 an acyclic saturated carbon skeleton
    • 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
    • C07C233/84Carboxylic 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 saturated carbon skeleton containing rings
    • 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
    • C07C235/34Carboxylic 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 atoms of the carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • 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
    • C07C235/52Carboxylic 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 having the nitrogen atom of at least one of the carboxamide groups bound to an acyclic carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • 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
    • C07C235/60Carboxylic 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 atoms of the carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • 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
    • C07C235/82Carboxylic 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 ring other than 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
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/28Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a non-condensed six-membered aromatic ring of the carbon skeleton
    • C07C237/40Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a non-condensed six-membered aromatic ring of the carbon skeleton having the nitrogen atom of the carboxamide group bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C279/00Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups
    • C07C279/04Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of guanidine groups bound to acyclic carbon atoms of a carbon skeleton
    • C07C279/14Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of guanidine groups bound to acyclic carbon atoms of a carbon skeleton being further substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/1072General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups
    • C07K1/1077General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups by covalent attachment of residues other than amino acids or peptide residues, e.g. sugars, polyols, fatty acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • C07K5/06043Leu-amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/0056Mouth soluble or dispersible forms; Suckable, eatable, chewable coherent forms; Forms rapidly disintegrating in the mouth; Lozenges; Lollipops; Bite capsules; Baked products; Baits or other oral forms for animals
    • 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
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated
    • 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
    • EFIXED CONSTRUCTIONS
    • E04BUILDING
    • E04FFINISHING WORK ON BUILDINGS, e.g. STAIRS, FLOORS
    • E04F11/00Stairways, ramps, or like structures; Balustrades; Handrails
    • E04F11/02Stairways; Layouts thereof
    • E04F11/104Treads
    • E04F2011/1046Miscellaneous features of treads not otherwise provided for
    • E04F2011/1048Miscellaneous features of treads not otherwise provided for with lighting means

Definitions

  • the present invention relates to methods and compositions for transporting active agents, and particularly biologically active agents, across cell membranes or lipid bilayers. These methods and compositions facilitate the delivery of an active agent to a target, such as the delivery of a pharmaceutical agent through an adverse environment to a particular location of the body.
  • Biologically active agents are particularly vulnerable to such barriers.
  • barriers are impeded by the body.
  • Subcutaneous, nasal or sublingual delivery to the circulatory system for many biologically active agents would be the route of choice for administration to animals if not for physical barriers such as the skin, lipid bi-layers, and various organ membranes that are relatively impermeable to certain biologically active agents, but one or more of which must be traversed before an agent delivered via these routes can reach the circulatory system.
  • delivery such as, for example, sublingual delivery may be impeded by chemical barriers such as the varying pH in the gastrointestinal (GI) tract and the presence of powerful digestive enzymes.
  • GI gastrointestinal
  • the initial focus of drug design is on the physiochemical properties of pharmaceutical compounds and particularly their therapeutic function.
  • the secondary design focus is on the need to deliver the active agent to its biological target(s). This is particularly true for drugs and other biologically active agents that are designed for oral administration to humans and other animals.
  • thousands of therapeutic compounds are discarded because no delivery systems are available to ensure that therapeutic titers of the compounds will reach the appropriate anatomical location or compartment(s) after administration and particularly oral administration.
  • many existing therapeutic agents are underutilized for their approved indications because of constraints on their mode(s) of administration.
  • many therapeutic agents could be effective for additional clinical indications beyond those for which they are already employed if there existed a practical methodology to deliver them in appropriate quantities to the appropriate biological targets.
  • the native state is usually described as the 3-dimensional state with the lowest free energy.
  • the signal peptides or the chaperonins facilitate the protein's ability to cross various cellular membranes until the protein reaches the appropriate organelle.
  • the chaperonin then separates from the protein or the signal peptide is cleaved from the protein, allowing the protein to fold to the native state. It is well known that the ability of the protein to transit cellular membranes is at least partly a consequence of being in a partially unfolded state.
  • diphtheria toxin and cholera toxin indicate that after diphtheria toxin binds to its cellular receptor, it is endocytosed, and while in this endocytic vesicle, it is exposed to an acidic pH environment.
  • the acidic pH induces a structural change in the toxin molecule which provides the driving force for membrane insertion and translocation to the cytosol.
  • Ramsay, G., Freire, E. Biochemistry, 1990, 29, 8677-8683 and Schon, A., Freire, E., Biochemistry, 1989, 28, 5019-5024 Similarly, cholera toxin undergoes a conformational change subsequent to endocytosis which allows the molecule to penetrate the nuclear membrane. See also, Morin, P. E., Diggs, D., Freire, E., Biochemistry, 1990, 29, 781-788.
  • Earlier designed delivery systems have used either an indirect or a direct approach to delivery.
  • the indirect approach seeks to protect the drug from a hostile environment. Examples are enteric coatings, liposomes, microspheres, microcapsules. See, colloidal drug delivery systems, 1994, ed. Jorg Freuter, Marcel Dekker, Inc.; U.S. Pat. No. 4,239,754; Patel et al. (1976), FEBS Letters, Vol. 62, pg. 60; and Hashimoto et al. (1979), Endocrinology Japan, Vol. 26, pg. 337.
  • the direct approach is based upon forming covalent linkages with the drug and a modifier, such as the creation of a prodrug.
  • a modifier such as the creation of a prodrug.
  • the linkage is usually designed to be broken under defined circumstances, e.g. pH changes or exposure to specific enzymes.
  • the covalent linkage of the drug to a modifier essentially creates a new molecule with new properties such as an altered log P value and/or as well as a new spatial configuration.
  • the new molecule has different solubility properties and is less susceptible to enzymatic digestion.
  • the present invention discloses methods for administering, by either the subcutaneous, nasal, or sublingual routes, a biologically active agent to a subject in need of such agent.
  • a first method includes the steps of:
  • the perturbant has a molecular weight between about 150 and about 600 daltons, and contains at least one hydrophilic moiety and at least one hydrophobic moiety.
  • the supramolecular complex comprises the perturbant non-covalently bound or complexed with the biologically active agent.
  • the biologically active agent does not form a microsphere after interacting with the perturbant.
  • compositions comprise a supramolecular complex including:
  • a complexing perturbant having a molecular weight ranging from about 150 to about 600 and having at least one hydrophilic moiety and at least one hydrophobic moiety wherein the perturbant is in an amount effective for subcutaneous, nasal, or sublingual delivery of the biologically active agent;
  • the intermediate state is conformationally between the native conformation state and denatured conformation state of the biologically active agent and the composition is not a microsphere.
  • an agent which is a mimetic which is capable of being administered, by the subcutaneous, nasal, or sublingual route, to a subject in need of such agent A biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to the native state and which is conformationally between the native state and the denatured state, is exposed to a complexing perturbant in an amount effective for subcutaneous, nasal, or sublingual delivery of such an agent to reversibly transform the biologically active agent to the intermediate conformational state and to form a transportable supramolecular complex.
  • the perturbant has a molecular weight between about 150 and about 600 daltons and at least one hydrophilic moiety and one hydrophobic moiety.
  • the supramolecular complex comprises the perturbant non-covalently complexed with the biologically active agent, and the biologically active agent does not form a microsphere with the perturbant. A mimetic of the supramolecular complex is prepared.
  • a method for preparing an agent which is capable of being administered either by the subcutaneous, nasal, or sublingual routes is provided.
  • a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to the native state and which is conformationally between the native and denatured states, is exposed to a perturbant to reversibly transform the biologically active agent to the intermediate state.
  • the perturbant is in a respective amount effective for subcutaneous, nasal, or sublingual delivery of the agent.
  • the agent a mimetic of the intermediate state, is prepared.
  • FIG. 1 is illustration of a native gradient gel of ⁇ -interferon (IFN) and a modified amino acid complexing perturbant.
  • IFN ⁇ -interferon
  • FIG. 2 is an illustration of a native gradient gel of ⁇ -interferon and a thermal condensate complexing perturbant.
  • FIG. 3 is a graphic illustration of guanidine hydrochloride (GuHCl) induced denaturation of ⁇ -interferon.
  • FIG. 4 is a graphic illustration of the concentration effect of GuHCl on ⁇ -interferon conformation.
  • FIG. 5 is a graphic illustration of the pH denaturation of ⁇ -interferon.
  • FIG. 6 is a graphic illustration of the pH denaturation of insulin.
  • FIGS. 7A and 7B are graphic illustrations of the reversibility of the circular dichroism spectrum of ⁇ -interferon.
  • FIG. 8 is a graphic illustration of the circular dichroism spectrum of ⁇ -interferon.
  • FIG. 9 is a graphic illustration of intrinsic tryptophan fluorescence of ⁇ -interferon and a complexing perturbant.
  • FIG. 10 is a graphic illustration of the differential scanning calorimetry of ⁇ -interferon and complexing perturbant.
  • FIGS. 11A and 11B are graphic illustrations of the reversibility of the transformation due to complexing perturbants.
  • FIG. 12 is a graphic illustration of the effect of complexing perturbant on ⁇ -interferon.
  • FIG. 13 is a graphic illustration of the concentration effect of complexing perturbant on ⁇ -interferon conformation.
  • FIG. 14 is a graphic illustration of the effect of complexing perturbant on ⁇ -interferon.
  • FIG. 15 is a graphic illustration of the Isothermal Titration Calorimetry of ⁇ -interferon and complexing perturbant.
  • FIG. 16 is a graphic illustration of the Isothermal Titration Calorimetry of ⁇ -interferon and complexing perturbant.
  • FIG. 17 is a graphic illustration of the effects of complexing perturbants on ⁇ -interferon.
  • FIG. 18 is a graphic illustration of the effect of the concentration of complexing perturbants on ⁇ -interferon.
  • FIG. 19 is a graphic illustration of the Isothermal Titration Calorimetry of ⁇ -interferon and complexing perturbant.
  • FIG. 20 is a graphic illustration of pancreatic inhibition assay with ⁇ -interferon and complexing perturbants.
  • FIG. 21 is a graphic illustration of the effect of DSC of heparin at pH 5.0.
  • FIG. 22 is a graphic illustration of the DSC of DPPC with perturbant compounds at several concentrations (units T M ,° C. vs. concentration).
  • FIG. 23 is a graphic illustration of the concentration effect of complexing perturbant compound L on the DPPC conformation.
  • FIG. 24 is a graphic illustration of the concentration effect of complexing perturbant compound L on rhGH conformation.
  • FIG. 25 is a graphic illustration of the concentration effect of complexing perturbant compound LI on rhGH conformation.
  • FIG. 26 is a graphic illustration of the concentration effect of complexing perturbant compound XI on rhGH conformation.
  • FIG. 27 is a graphic illustration of the differential light scattering of perturbant compound L in a 10 mM phosphate buffer at pH 7.0.
  • FIG. 28 is a graphic illustration of the results of subcutaneous injection of rhGH composition in rats.
  • FIG. 29 is a graphic illustration of the results of Sublingual (SL), intranasal (IN), and intracolonic (IC) dosing of rhGH in rats.
  • FIG. 30 is a graphic illustration of the results of intracolonic dosing of heparin with compound XXXI carrier.
  • the present methods effect active agent delivery by creating a reversibly non-covalently complexed supramolecule from the active agent and an amount of complexing perturbant appropriate for the route of delivery.
  • the three-dimensional structure or conformation of the active agent is changed, but the chemical composition of the active agent molecule is not altered.
  • This alteration in structure provides the active agent with the appropriate properties, such as, for example, solubility (log P) to cross or penetrate a physical or chemical barrier, membrane, or lipid bilayer, to resist enzymatic degradation and the like.
  • Crossing refers to transport from one side of the cell membrane or lipid bilayer to the opposite side (i.e.
  • the perturbed intermediate state of the active agent or the supramolecular complex itself can be used as a template for the preparation of mimetics which, accordingly, could be delivered to a target by the appropriate route, i.e., subcutaneous, nasal, or sublingual.
  • an active agent After crossing the cell membrane or lipid bilayer, an active agent has biological activity and bioavailability, either by restoration to the native state or by retaining biological activity or bioavailability acquired in the intermediate state.
  • the mimetic acts similarly after crossing the cell membrane or lipid bilayer.
  • the native conformational state of an active agent is typically described as the three dimensional state with the lowest free energy ( ⁇ G). It is the state in which the active agent typically possesses the full complement of activity ascribed to the agent, such as the full complement of biological activity ascribed to a biologically active agent.
  • the denatured conformational state is the state in which the active agent has no secondary or tertiary structure.
  • Intermediate conformational states exist between the native and denatured states.
  • a particular active agent may have one or more intermediate states.
  • the intermediate state achieved by the present invention is structurally and energetically distinct from both the native and denatured states.
  • Active agents useful in the present invention must be transformable from their native conformational state to an intermediate conformational state which can be administered by the route of choice and back to their native state, i.e. reversibly transformable, so that when the active agent reaches its target, such as when a delivered drug reaches the circulatory system, the active agent retains, regains, or acquires a biologically, pharmacologically, or therapeutically significant complement of its desired biological activity.
  • the ⁇ G of the intermediate state ranges from about ⁇ 20 Kcal/mole to about 20 Kcal/mole, and most preferably, it ranges from about ⁇ 10 Kcal/mole to about 10 Kcal/mole, all relative to the native state.
  • the intermediate state has significant secondary structure, significant compactness due to the presence of a sizable hydrophobic core, and a tertiary structure reminiscent of the native fold but without necessarily exhibiting the packing of the native state.
  • the difference in free energy ( ⁇ G) between the intermediate state and the native state is relatively small.
  • the equilibrium constant between the native and the transportable, reversible intermediate state(s) is close to unity (depending upon experimental conditions).
  • Intermediate states can be confirmed by, for example, differential scanning calorimetry (DSC), isothermal titration calorimetry (ITC), native gradient gels, NMR, fluorescence, and the like.
  • ⁇ G 0 ⁇ H 0 ( T R ) ⁇ T ⁇ S 0 ( T R )+ ⁇ Cp 0 (( T ⁇ T R ) ⁇ T In ( T/T R )) (1)
  • T is the temperature
  • T R is a reference temperature
  • ⁇ H 0 (T R ) and T ⁇ S 0 (T R ) are the relative enthalpy and entropy of this state at the reference temperature
  • ⁇ Cp 0 is the relative heat capacity of this state. It is convenient to chose the native state as the reference state to express all relative thermodynamic parameters.
  • Active agents suitable for use in the present invention include biologically active agents and chemically active agents, including, but not limited to, fragrances, as well as other active agents such as, for example, cosmetics.
  • Bioly active agents include, but are not limited to, pesticides, pharmacological agents, and therapeutic agents.
  • biologically active agents suitable for use in the present invention include, but are not limited to, peptides, and particularly small peptides; hormones, and particularly hormones which by themselves do not or only pass slowly through the gastro-intestinal mucosa and/or are susceptible to chemical cleavage by acids and enzymes in the gastrointestinal tract; polysaccharides, and particularly mixtures of muco-polysaccharides; carbohydrates; lipids; or any combination thereof.
  • Further examples include, but are not limited to, human growth hormones; bovine growth hormones; growth releasing hormones; interferons; interleukin-1; insulin; heparin, and particularly low molecular weight heparin; calcitonin; erythropoietin; atrial naturetic factor; antigens; monoclonal antibodies; somatostatin; adrenocorticotropin, gonadotropin releasing hormone; oxytocin; vasopressin; cromolyn sodium (sodium or disodium chromoglycate); vancomycin; desferrioxamine (DFO); anti-microbials, including, but not limited to anti-fungal agents; or any combination thereof.
  • human growth hormones bovine growth hormones
  • growth releasing hormones interferons
  • interleukin-1 insulin
  • insulin heparin, and particularly low molecular weight heparin
  • calcitonin erythropoietin
  • atrial naturetic factor
  • compositions of the present invention may combine one or more active agents.
  • Perturbants serve two purposes in the present invention.
  • the active agent is contacted with an amount of perturbant which reversibly transforms the active agent from the native state to the intermediate state suitable for administration.
  • the perturbant in the appropriate amount, non-covalently complexes with the active agent to form a supramolecular complex which can be administered by a selected route.
  • This supramolecular complex can be used as a template for the design of a mimetic or can be used as a delivery composition itself.
  • the perturbant in effect, fixes the active agent in the transportable intermediate state.
  • the perturbant can be released from the supramolecular complex, such as by dilution in the circulatory system, so that the active agent can return to the native state appropriate for the route of delivery.
  • these perturbants have at least one hydrophilic (i.e. readily soluble in water, such as for example, a carboxylate group) and at least one hydrophobic moiety (i.e. readily soluble in an organic solvent such as, for example, a benzene group), and have a molecular weight ranging from about 150 to about 600 daltons and most preferably from about 200 to about 500 daltons.
  • Complexing perturbant compounds include, but are not limited to proteinoids including linear, non-linear, and cyclic proteinoids; modified (acylated or sulfonated) amino acids, poly amino acids, and peptides; modified amino acid, poly amino acid, or peptide derivatives (ketones or aldehydes); diketopiperazine/amino acid constructs; carboxylic acids; and various other perturbants discussed below.
  • non-covalent complexing may be effected by intermolecular forces including but not limited to, hydrogen bonding, hydrophilic interactions, electrostatic interactions, and Van der Waals interactions.
  • intermolecular forces including but not limited to, hydrogen bonding, hydrophilic interactions, electrostatic interactions, and Van der Waals interactions.
  • the dissociation constant K d is the reciprocal of K a .
  • Protein unfolding can be described according to the equilibrium that exists between its various conformational states, e.g. N ⁇ ⁇ k 1 ⁇ I ⁇ D k 2 ( 7 )
  • N is the native state
  • I is the intermediate state(s)
  • D is the denatured state
  • k 1 and k 2 are the respective rate constants.
  • the complexing of the perturbant with the active agent must be strong enough to insure delivery of the drug either to the systemic circulation and/or to the target(s), but not so strong so that disengagement of the perturbant will not occur in a timely manner to allow the active agent to renature if necessary to produce the desired effect(s).
  • amounts of perturbants appropriate for the route of delivery reversibly transform the active agent to the intermediate state so that the conformation of that state can be used as a template for the preparation of mimetics.
  • Perturbants for this purpose need not, but may, complex with the active agent. Therefore, in addition to the complexing perturbants discussed above, perturbants that change the pH of the active agent or its environment, such as for example, strong acids or strong bases; detergents; perturbants that change the ionic strength of the active agent or its environment; other agents such as for example, guanidine hydrochloride; and temperature can be used to transform the active agent. Either the supramolecular complex or the reversible intermediate state can be used as a template for mimetic design.
  • Amino acids are the basic materials used to prepare many of the complexing perturbants useful in the present invention.
  • An amino acid is any carboxylic acid having at least one free amine group and includes naturally occurring and synthetic amino acids.
  • the preferred amino acids for use in the present invention are ⁇ -amino acids, and most preferably are naturally occurring ⁇ -amino acids.
  • Many amino acids and amino acid esters are readily available from a number of commercial sources such as Aldrich Chemical Co. (Milwaukee, Wis., U.S.A.); Sigma Chemical Co. (St. Louis, Mo., U.S.A.); and Fluka Chemical Corp. (Ronkonkoma, N.Y., U.S.A.).
  • amino acids suitable for use in the present invention are generally of the formula
  • R 1 is hydrogen, C 1 -C 4 alkyl, or C 2 -C 4 alkenyl
  • R 2 is C 1 -C 24 alkyl, C 2 -C 24 alkenyl, C 3 -C 10 cycloalkyl, C 3 -C 10 cycloalkenyl, phenyl, naphthyl, (C 1 -C 10 alkyl) phenyl, (C 2 -C 10 alkenyl) phenyl, (C 1 -C 10 alkyl) naphthyl, (C 2 -C 10 alkenyl) naphthyl, phenyl (C 1 -C 10 alkyl), phenyl (C 2 -C 10 alkenyl), naphthyl (C 1 -C 10 alkyl), or naphthyl (C 2 -C 10 alkenyl);
  • R 2 being optionally substituted with C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 1 -C 4 alkoxy, —OH, —SH, —CO 2 R 3 , C 3 -C 10 cycloalkyl, C 3 -C 10 cycloalkenyl, heterocycle having 3-10 ring atoms wherein the hetero atom is one or more of N, O, S, or any combination thereof, aryl, (C 1 -C 10 alk)aryl, ar(C 1 -C 10 alkyl) or any combination thereof;
  • R 2 being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof;
  • R 3 is hydrogen, C 1 -C 4 alkyl, or C 2 -C 4 alkenyl.
  • amino acids or components of a peptide are alanine, arginine, asparagine, aspartic acid, citrulline, cysteine, cystine, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, ornithine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, hydroxy proline, ⁇ -carboxyglutamate, phenylglycine, or O-phosphoserine.
  • the preferred amino acids are arginine, leucine, lysine, phenylalanine, tyrosine, tryptophan, valine, and phenylglycine.
  • the preferred non-naturally occurring amino acids for use in the present invention are ⁇ -alanine, ⁇ -amino butyric acid, ⁇ -amino butyric acid, ⁇ -(aminophenyl) butyric acid, ⁇ -amino isobutyric acid, citrulline, ⁇ -amino caproic acid, 7-amino heptanoic acid, ⁇ -aspartic acid, aminobenzoic acid, aminophenyl acetic acid, aminophenyl butyric acid, ⁇ -glutamic acid, cysteine (ACM), ⁇ -lysine, ⁇ -lysine (A-Fmoc), methionine sulfone, norleucine, norvaline, ornithine, d-ornithine, p-nitro-phenylalanine, hydroxy proline, 1,2,3,4,-tetrahydroisoquinoline-3-carboxylic acid, and thioproline.
  • Poly amino acids are either peptides or two or more amino acids linked by a bond formed by other groups which can be linked, e.g., an ester, anhydride or an anhydride linkage. Special mention is made of non-naturally occurring poly amino acids and particularly non-naturally occurring hetero-poly amino acids, i.e. of mixed amino acids.
  • Peptides are two or more amino acids joined by a peptide bond. Peptides can vary in length from di-peptides with two amino acids to polypeptides with several hundred amino acids. See, Walker, Chambers Biological Dictionary, Cambridge, England: Chambers Cambridge, 1989, page 215. Special mention is made of non-naturally occurring peptides and particularly non-naturally occurring peptides of mixed amino acids. Special mention is also made of di-peptides tri-peptides, tetra-peptides, and penta-peptides, and particularly, the preferred peptides are di-peptides and tri-peptides. Peptides can be homo- or hetero-peptides and can include natural amino acids, synthetic amino acids, or any combination thereof.
  • Proteinoids are artificial polymers of amino acids.
  • the proteinoids preferably are prepared from mixtures of amino acids.
  • Preferred proteinoids are condensation polymers, and most preferably, are thermal condensation polymers. These polymers may be directed or random polymers.
  • Proteinoids can be linear, branched, or cyclical, and certain proteinoids can be units of other linear, branched, or cyclical proteinoids.
  • Diketopiperazines are six member ring compounds.
  • the ring includes two nitrogen atoms and is substituted at two carbons with two oxygen atoms.
  • the carbonyl groups are at the 1 and 4 ring positions. These rings can be optionally, and most often are, further substituted.
  • Diketopiperazine ring systems may be generated during thermal polymerization or condensation of amino acids or amino acid derivatives. (Gyore, J; Ecet M. Proceedings Fourth ICTA ( Thermal Analysis ), 1974, 2, 387-394 (1974)). These six membered ring systems were presumably generated by intra-molecular cyclization of the dimer prior to further chain growth or directly from a linear peptide (Reddy, A. V., Int. J. Peptide Protein Res., 40, 472-476 (1992); Mazurov, A. A. et al., Int. J. Peptide Protein Res., 42, 14-19 (1993)).
  • Diketopiperazines can also be formed by cyclodimerization of amino acid ester derivatives as described by Katchalski et al., J. Amer. Chem. Soc., 68, 879-880 (1946), by cyclization of dipeptide ester derivatives, or by thermal dehydration of amino acid derivatives and high boiling solvents as described by Kopple et al., J. Org. Chem., 33 (2), 862-864 (1968).
  • Diketopiperazines typically are formed from ⁇ -amino acids.
  • the ⁇ -amino acids of which the diketopiperazines are derived are glutamic acid, aspartic acid, tyrosine, phenylalanine, and optical isomers of any of the foregoing.
  • R 4 , R 5 , R 6 , and R 7 independently are hydrogen, C 1 -C 24 alkyl, C 1 -C 24 alkenyl, phenyl, naphthyl, (C 1 -C 10 alkyl)phenyl, (C 1 -C 10 alkenyl)phenyl, (C 1 -C 10 alkyl)naphthyl, (C 1 -C 10 alkenyl)naphthyl, phenyl (C 1 -C 10 alkyl), phenyl(C 1 -C 10 alkenyl), naphthyl (C 1 -C 10 alkyl), and naphthyl (C 1 -C 10 alkenyl); any of R 4 , R 5 , R 6 , and R 7 independently may optionally be substituted with C 1 -C 4 alkyl, C 1 -C 4 alkenyl, C 1 -C 4 alkoxy, —OH, —SH
  • the phenyl or naphthyl groups may optionally be substituted. Suitable, but non-limiting, examples of substituents are C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C 1 -C 6 alkoxy, —OH, —SH, or CO 2 R 9 wherein R 9 is hydrogen, C 1 -C 6 alkyl, or C 1 -C 6 alkenyl.
  • R 6 and R 7 independently are hydrogen, C 1 -C 4 alkyl or C 1 -C 4 alkenyl.
  • diketopiperazines which are preferred complexing perturbants. These diketopiperazines include the unsubstituted diketopiperazine in which R 4 , R 5 , R 6 , and R 7 are hydrogen, and diketopiperazines which are substituted at one or both of the nitrogen atoms in the ring, i.e. mono or di-N-substituted. Special mention is made of the N-substituted diketopiperazine wherein one or both of the nitrogen atoms is substituted with a methyl group.
  • R 10 and R 11 independently are hydrogen, C 1 -C 24 alkyl, C 1 -C 24 alkenyl, phenyl, naphthyl, (C 1 -C 10 alkyl)phenyl, (C 1 -C 10 alkenyl)phenyl, (C 1 -C 10 alkyl)naphthyl, (C 1 -C 10 alkenyl)naphthyl, phenyl (C 1 -C 10 alkyl), phenyl(C 1 -C 10 alkenyl), naphthyl (C 1 -C 10 alkyl), and naphthyl (C 1 -C 10 alkenyl); but both R 10 and R 11 can not be hydrogen; either or both R 10 or R 11 independently may optionally be substituted with C 1 -C 4 alkyl, C 1 -C 4 alkenyl, C 1 -C 4 alkoxy, —OH, —SH, and —CO 2 R
  • the phenyl or naphthyl groups may optionally be substituted. Suitable, but non-limiting, examples of substituents are C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C 1 -C 6 alkoxy, —OH, —SH, or CO 2 R 13 wherein R 13 is hydrogen, C 1 -C 6 alkyl, or C 1 -C 6 alkenyl.
  • R 10 or R 11 is hydrogen
  • the diketopiperazine is mono-carbon-(C)-substituted.
  • neither R 10 nor R 11 is hydrogen
  • the diketopiperazine is di-carbon-(C)-substituted.
  • R 10 , R 11 , or both R 10 and R 11 contain at least one functional group, a functional group being a non-hydrocarbon portion responsible for characteristic reactions of the molecule.
  • Simple functional groups are heteroatoms including, but not limited to halogens, oxygen, sulfur, nitrogen, and the like, attached to, the carbon of an alkyl group by a single or multiple bond.
  • Other functional groups include, but are not limited to, for example, hydroxyl groups, carboxyl groups, amide groups, amine groups, substituted amine groups, and the like.
  • Preferred diketopiperazines are those which are substituted at one or two of the carbons of the ring with a functional group that includes at least one carboxyl functionality.
  • Diketopiperazines may also be polymerized with additional amino acids to form constructs of at least one amino acid or an ester or an amide thereof and at least one diketopiperazine, preferably covalently bonded to one another.
  • one or more of the R groups must contain at least one functional group, a functional group being a non-hydrocarbon portion responsible for characteristic reactions of the molecule.
  • Simple functional groups are heteroatoms including, but not limited to halogens, oxygen, sulfur, nitrogen, and the like, attached to, the carbon of an alkyl group by a single or multiple bond.
  • Other functional groups include, but are not limited to, for example, hydroxyl groups, carboxyl groups, amide groups, amine groups, substituted amine groups, and the like.
  • diketopiperazines which are preferred components of the amino acids/diketopiperazine perturbants of the present invention.
  • preferred diketopiperazines are those which are substituted at one or two of the carbons of the ring and preferably are substituted with a functional group that includes at least one carboxyl functionality.
  • the diketopiperazines in the amino acids/diketopiperazine perturbants are prepared from trifunctional amino acids such as L-glutamic acid and L-aspartic acid which cyclize to form diketopiperazines.
  • the diketopiperazines can generate a bis-carboxylic acid platform which can be further condensed with other amino acids to form the perturbant.
  • the diketopiperazine will react and covalently bond with one or more of the amino acids through the functional group(s) of the R groups of the diketopiperazines.
  • the amino acids/diketopiperazine perturbants may include one or more of the same or different amino acids as well as one or more of the same or different diketopiperazines as described above.
  • Modified amino acids, poly amino acids or peptides are either acylated or sulfonated and include amino acid amides and sulfonamides.
  • Modified amino acids are typically prepared by modifying the amino acid or an ester thereof. Many of these compounds are prepared by acylation or sulfonation with agents having the formula
  • R 4 is the appropriate radical to yield the modification indicated in the final product
  • X is a leaving group.
  • Typical leaving groups include, but are not limited t, halogens such as, for example, chlorine, bromine, and iodine. Additionally, the corresponding anhydrides are modifying agents.
  • Ar is a substituted or unsubstituted phenyl or naphthyl
  • R 14 has the formula
  • R 15 is C 1 to C 24 alkyl, C 1 to C 24 alkenyl, phenyl, naphthyl, (C 1 to C 10 alkyl) phenyl, (C 1 to C 10 alkenyl) phenyl, (C 1 to C 10 alkyl) naphthyl, (C 1 to C 10 alkenyl) naphthyl, phenyl (C 1 to C 10 alkyl), phenyl (C 1 to C 10 alkenyl), naphthyl (C 1 to C 10 alkyl) and naphthyl (C 1 to C 10 alkenyl);
  • R 15 is optionally substituted with C 1 to C 4 alkyl, C 1 to C 4 alkenyl, C 1 to C 4 alkoxy, —OH, —SH and —CO 2 R 5 , cycloalkyl, cycloalkenyl, heterocyclic alkyl, alkaryl, heteroaryl, heteroalkaryl, or any combination thereof;
  • R 17 is hydrogen, C 1 to C 4 alkyl or C 1 to C 4 alkenyl
  • R 15 is optionally interrupted by oxygen, nitrogen, sulfur or any combination thereof.
  • R 16 is hydrogen, C 1 to C 4 alkyl or C 1 to C 4 alkenyl.
  • R 19 is hydrogen, C 1 -C 4 alkyl, or C 2 -C 4 alkenyl
  • R 20 is C 1 -C 24 alkyl, C 2 -C 24 alkenyl, C 3 -C 10 cycloalkyl, C 3 -C 10 cycloalkenyl, phenyl, naphthyl, (C 1 -C 10 alkyl) phenyl, (C 2 -C 10 alkenyl) phenyl, (C 1 -C 10 alkyl) naphthyl, (C 2 -C 10 alkenyl) naphthyl, phenyl (C 1 -C 10 alkyl), phenyl (C 2 -C 10 alkenyl), naphthyl (C 1 -C 10 alkyl) or naphthyl (C 2 -C 10 alkenyl);
  • R 20 being optionally substituted with C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 1 -C 4 alkoxy, —OH, —SH, —CO 2 R 22 , C 3 -C 10 cycloalkyl, C 3 -C 10 cycloalkenyl, heterocycle having 3-10 ring atoms wherein the hetero atom is one or more of N, O, S or any combination thereof, aryl, (C 1 -C 10 alk)aryl, ar(C 1 -C 10 alkyl), or any combination thereof;
  • R 20 being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof;
  • R 22 is hydrogen, C 1 -C 4 alkyl, or C 2 -C 4 alkenyl.
  • Some preferred acylated amino acids include salicyloyl phenylalanine, and the compounds having the formulas: A Compound n m X XXXVIIA 0 0 4-Cl XXXVIIIA 3 0 H XXIXA 3 1 4-CH 3 XLA 3 1 2-F XLIA 3 1 2-CH 3 XLIIA 3 0 3-CF 3 XLIIIA 3 4 H XLIVA 3 0 3-Cl XLVA 3 0 3-F XLVIA 3 0 3-CH 3 XLVIIA 0 0 2-CF 3 XLVIIIA 1 2 H XLIXA 3 2 2-F LA 3 0 3,4-OCH 2 O— LIA 3 0 2-COOH LIIA 1 0 2-OH LIIIA 3 0 2,6-dihydroxy LIVA 2 0 2-OH LVA 0 0 2,4-difluoro LVIA 2 0 2,6-dihydroxy LVIIA 0 0 0
  • organic acid compounds, and their salts, having an aromatic amide group, having a hydroxy group substituted in the ortho position on the aromatic ring, and a lipophilic chain with from about 4 carbon atoms to about 20 atoms in the chain are also useful as perturbants.
  • the lipophilic chain can have from 5 to 20 carbon atoms.
  • Perturbants which are also useful also include those having the formula
  • Ar is a substituted or unsubstituted phenyl or naphthyl
  • R 7 is selected from the group consisting of C 4 to C 20 alkyl, C 4 to C 20 alkenyl, phenyl, naphthyl, (C 1 to C 10 alkyl) phenyl, (C 1 to C 10 alkenyl) phenyl, (C 1 to C 10 alkyl) naphthyl, (C 1 to C 10 alkenyl) naphthyl, phenyl (C 1 to C 10 alkyl), phenyl (C 1 to C 10 alkenyl), naphthyl (C 1 to C 10 alkyl), and naphthyl (C 1 to C 10 alkenyl);
  • R 8 is selected from the group consisting of hydrogen, C 1 to C 4 alkyl, C 1 to C 4 alkenyl, hydroxy, and C 1 to C 4 alkoxy;
  • R 7 is optionally substituted with C 1 to C 4 alkyl, C 1 to C 4 alkenyl, C 1 to C 4 alkoxy, —OH, —SH and —CO 2 R 9 or any combination thereof;
  • R 9 is hydrogen, C 1 to C 4 alkyl or C 1 to C 4 alkenyl
  • R 7 is optionally interrupted by oxygen, nitrogen, sulfur or any combination thereof;
  • the preferred R 6 groups are of C 4 to C 20 alkyl and C 4 to C 20 alkenyl.
  • the most preferred R 6 groups are C 5 to C 20 alkyl and C 5 to C 20 alkenyl.
  • a preferred carrier compound can have the formula
  • A is Try, Leu, Arg, Trp, or Cit
  • A is Try, Arg, Trp or Cit; A is acylated at 2 or more functional groups.
  • Preferred compounds also include those wherein A is Try; A is Tyr and is acylated at 2 functional groups; A is Leu; A is Arg; A is Arg and is acylated at 2 functional groups; A is Trp; A is Trp and is acylated at 2 functional groups; A is Cit; and A is Cit and is acylated at 2 functional groups.
  • A is Arg or Leu
  • A is Arg, A is optionally acylated at 2 or more functional groups;
  • A is Leu or phenylglycine
  • A is phenylglycine
  • A is phenylglycine
  • Acylated amino acids may be prepared by reacting single amino acids, mixtures of two or more amino acids, or amino acid esters with an amine modifying agent which reacts with free amino moieties present in the amino acids to form amides.
  • Suitable, but non-limiting, examples of acylating agents useful in preparing acylated amino acids include acid chloride acylating agents having the formula
  • R 23 an appropriate group for the modified amino acid being prepared, such as, but not limited to, alkyl, alkenyl, cycloalkyl, or aromatic, and particularly methyl, ethyl, cyclohexyl, cyclophenyl, phenyl, or benzyl, and X is a leaving group.
  • Typical leaving groups include, but are not limited to, halogens such as chlorine, bromine and iodine.
  • acylating agents include, but are not limited to, acyl halides including, but not limited to, acetyl chloride, propyl chloride, cyclohexanoyl chloride, cyclopentanoyl chloride, and cycloheptanoyl chloride, benzoyl chloride, hippuryl chloride and the like; and anhydrides, such as acetic anhydride, propyl anhydride, cyclohexanoic anhydride, benzoic anhydride, hippuric anhydride and the like.
  • acyl halides including, but not limited to, acetyl chloride, propyl chloride, cyclohexanoyl chloride, cyclopentanoyl chloride, and cycloheptanoyl chloride, benzoyl chloride, hippuryl chloride and the like
  • anhydrides such as acetic anhydride, propyl anhydride, cyclohexanoic anhydride
  • Preferred acylating agents include benzoyl chloride, hippuryl chloride, acetyl chloride, cyclohexanoyl chloride, cyclopentanoyl chloride, and cycloheptanoyl chloride.
  • the amine groups can also be modified by the reaction of a carboxylic acid with coupling agents such as the carbodiimide derivatives of amino acids, particularly hydrophilic amino acids such as phenylalanine, tryptophan, and tyrosine. Further examples include dicyclohexylcarbodiimide and the like.
  • the amino acid is multifunctional, i.e. has more than one —OH, —NH 2 or —SH group, then it may optionally be acylated at one or more functional groups to form, for example, an ester, amide, or thioester linkage.
  • the amino acids are dissolved in an aqueous alkaline solution of a metal hydroxide, e.g., sodium or potassium hydroxide and the acylating agent added.
  • the reaction time can range from about 1 hour and about 4 hours, preferably about 2-2.5 hours.
  • the temperature of the mixture is maintained at a temperature generally ranging between about 5° C. and about 70° C., preferably between about 10° C. and about 50° C.
  • the amount of alkali employed per equivalent of NH 2 groups in the amino acids generally ranges between about 1.25 moles and about 3 moles, and is preferably between about 1.5 moles and about 2.25 moles per equivalent of NH 2 .
  • the pH of the reaction solution generally ranges between about pH 8 and about pH 13, and is preferably between about pH 10 and about pH 12.
  • the amount of amino modifying agent employed in relation to the quantity of amino acids is based on the moles of total free NH 2 in the amino acids. In general, the amino modifying agent is employed in an amount ranging between about 0.5 and about 2.5 mole equivalents, preferably between about 0.75 and about 1.25 equivalents, per molar equivalent of total NH 2 groups in the amino acids.
  • the modified amino acid formation reaction is quenched by adjusting the pH of the mixture with a suitable acid, e.g., concentrated hydrochloric acid, until the pH reaches between about 2 and about 3.
  • a suitable acid e.g., concentrated hydrochloric acid
  • the mixture separates on standing at room temperature to form a transparent upper layer and a white or off-white precipitate.
  • the upper layer is discarded, and modified amino acids are collected by filtration or decantation.
  • the crude modified amino acids are then mixed with water. Insoluble materials are removed by filtration and the filtrate is dried in vacuo.
  • the yield of modified amino acids generally ranges between about 30 and about 60%, and usually about 45%.
  • the present invention also contemplates amino acids which have been modified by multiple acylation, e.g., diacylation or triacylation.
  • amino acid esters such as, for example benzyl, methyl, or ethyl esters of amino acid compounds
  • the amino acid ester dissolved in a suitable organic solvent such as dimethylformamide, pyridine, or tetrahydrofuran is reacted with the appropriate amino modifying agent at a temperature ranging between 5° C. and about 70° C., preferably about 25° C., for a period ranging between about 7 and about 24 hours.
  • the amount of amino acid modifying agent used relative to the amino acid ester is the same as described above for amino acids.
  • This reaction may be carried out with or without a base such as, for example, triethylamine or diisopropylethylamine.
  • amino acid esters or amides are the starting materials, they are dissolved in a suitable organic solvent such as dimethylformamide or pyridine, are reacted with the amino modifying agent at a temperature ranging between about 5° C. and about 70° C., preferably about 25° C., for a period ranging between about 7 and about 24 hours.
  • a suitable organic solvent such as dimethylformamide or pyridine
  • the amount of amino modifying agents used relative to the amino acid esters are the same as described above for amino acids.
  • the reaction solvent is removed under negative pressure and optionally the ester or amide functionality can be removed by hydrolyzing the modified amino acid ester with a suitable alkaline solution, e.g., 1N sodium hydroxide, at a temperature ranging between about 50° C. and about 80° C., preferably about 70° C., for a period of time sufficient to hydrolyze off the ester group and form the modified amino acid having a free carboxyl group.
  • the hydrolysis mixture is then cooled to room temperature and acidified, e.g., with an aqueous 25% hydrochloric acid solution, to a pH ranging between about 2 and about 2.5.
  • the modified amino acid precipitates out of solution and is recovered by conventional means such as filtration or decantation.
  • the modified amino acids may be purified by acid precipitation, recrystallization or by fractionation on solid column supports. Fractionation may be performed on a suitable solid column supports such as silica gel, alumina, using solvent mixtures such as acetic acid/butanol/water as the mobile phase; reverse phase column supports using trifluoroacetic acid/acetonitrile mixtures as the mobile phase; and ion exchange chromatography using water as the mobile phase.
  • the modified amino acids may also be purified by extraction with a lower alcohol such as methanol, butanol, or isopropanol to remove impurities such as inorganic salts.
  • the modified amino acids generally are soluble in alkaline aqueous solution (pH ⁇ 9.0); partially soluble in ethanol, n-butanol and 1:1 (v/v) toluene/ethanol solution and insoluble in neutral water.
  • the alkali metal salts e.g., the sodium salt of the derivatized amino acids are generally soluble in water at about a pH of 6-8.
  • poly amino acids or peptides one or more of the amino acids may be modified (acylated).
  • Modified poly amino acids and peptides may include one or more acylated amino acid(s).
  • linear modified poly amino acids and peptides will generally include only one acylated amino acid, other poly amino acid and peptide configurations can include more than one acylated amino acid.
  • Poly amino acids and peptides can be polymerized with the acylated amino acid(s) or can be acylated after polymerization.
  • a and B independently are Arg or Leu.
  • Sulfonated modified amino acids, poly amino acids, and peptides are modified by sulfonating at least one free amine group with a sulfonating agent which reacts with at least one of the free amine groups present.
  • Ar is a substituted or unsubstituted phenyl or naphthyl
  • R 25 is C 1 to C 24 alkyl, C 1 to C 24 alkenyl, phenyl, naphthyl, (C 1 to C 10 alkyl) phenyl, (C 1 to C 10 alkenyl) phenyl, (C 1 to C 10 alkyl) naphthyl, (C 1 to C 10 alkenyl) naphthyl, phenyl (C 1 to C 10 alkyl), phenyl (C 1 to C 10 alkenyl), naphthyl (C 1 to C 10 alkyl) and naphthyl (C 1 to C 10 alkenyl);
  • R 25 is optionally substituted with C 1 to C 4 alkyl, C 1 to C 4 alkenyl, C 1 to C 4 alkoxy, —OH, —SH and —CO 2 R 27 or any combination thereof;
  • R 27 is hydrogen, C 1 to C 4 alkyl or C 1 to C 4 alkenyl
  • R 25 is optionally interrupted by oxygen, nitrogen, sulfur or any combination thereof.
  • R 26 is hydrogen, C 1 to C 4 alkyl or C 1 to C 4 alkenyl.
  • Suitable, but non-limiting, examples of sulfonating agents useful in preparing sulfonated amino acids include sulfonating agents having the formula R 28 —SO 2 —X wherein R 28 is an appropriate group for the modified amino acid being prepared such as, but not limited to, alkyl, alkenyl, cycloalkyl, or aromatics and X is a leaving group as described above.
  • R 28 is an appropriate group for the modified amino acid being prepared such as, but not limited to, alkyl, alkenyl, cycloalkyl, or aromatics and X is a leaving group as described above.
  • a sulfonating agent is benzene sulfonyl chloride.
  • Modified poly amino acids and peptides may include one or more sulfonated amino acid(s). Although linear modified poly amino acids and peptides used generally include only one sulfonated amino acid, other poly amino acid and peptide configurations can include more than one sulfonated amino acid. Poly amino acids and peptides can be polymerized with the sulfonated amino acid(s) or can be sulfonated after polymerization.
  • Modified amino acid, polyamino acid, or peptide derivatives are amino acids, poly amino acids, or peptides which have had at least one acyl-terminus converted to an aldehyde or a ketone, and are acylated at at least one free amine group, with an acylating agent which reacts with at least one of the free amine groups present.
  • Amino acid, poly amino acid, or peptide derivatives can be readily prepared by reduction of amino acid esters or peptide esters with an appropriate reducing agent.
  • amino acid, poly amino acid, or peptide aldehydes can be prepared as described in an article by R. Chen et al., Biochemistry, 1979, 18, 921-926.
  • Amino acid, poly amino acid, or peptide ketones can be prepared by the procedure described in Organic Syntheses, Col. Vol. IV, Wiley, (1963), page 5. Acylation is discussed above.
  • the derivatives may be prepared by reacting a single amino acid, poly amino acid, or peptide derivative or mixtures of two or more amino acid or peptide derivatives, with an acylating agent or an amine modifying agent which reacts with free amino moieties present in the derivatives to form amides.
  • the amino acid, poly amino acid, or peptide can be modified and subsequently derivatized, derivatized and subsequently modified, or simultaneously modified and derivatized.
  • Protecting groups may be used to avoid unwanted side reactions as would be known to those skilled in the art.
  • one or more of the amino acid may be derivatized (an aldehyde or a ketone) and/or modified, (acylated) but there must be at least one derivative and at least one modification.
  • carboxylic acids and salts of these carboxylic acids may be used as complexing perturbants. These carboxylic acids have the formula:
  • R 29 is C 1 to C 24 alkyl, C 2 to C 24 alkenyl, C 3 to C 10 cycloalkyl, C 3 to C 10 cycloalkenyl, phenyl, naphthyl, (C 1 to C 10 alkyl)phenyl, (C 2 to C 10 alkenyl)phenyl, (C 1 to C 10 alkyl)naphthyl, (C 2 to C 10 alkenyl)naphthyl, phenyl(C 1 to C 10 alkyl), phenyl(C 2 to C 10 alkenyl), naphthyl(C 1 to C 10 alkyl) and naphthyl(C 2 to C 10 alkenyl);
  • R 29 being optionally substituted with C 1 to C 10 alkyl, C 2 to C 10 alkenyl, C 1 to C 4 alkoxy, —OH, —SH, —CO 2 R 30 , C 3 to C 10 cycloalkyl, C 3 to C 10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C 1 to C 10 alk)aryl, aryl(C 1 to C 10 )alkyl, or any combination thereof;
  • R 29 being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof.
  • R 30 is hydrogen, C 1 to C 4 alkyl or C 2 to C 4 alkenyl.
  • the preferred carboxylic acids are cyclohexanecarboxylic acid, cyclopentanecarboxylic acid, cycloheptanecarboxylic acid, hexanoic acid, 3-cyclohexanepropanoic acid, methylcyclohexanecarboxylic acid, 1,2-cyclohexanedicarboxylic acid, 1,3-cyclohexanedicarboxylic acid, 1,4-cyclohexanedicarboxylic acid, 1-adamantanecarboxylic acid, phenylpropanoic acid, adipic acid, cyclohexanepentanoic acid, cyclohexanebutanoic acid, pentylcyclohexanoic acid, 2-cyclopentanehexanoic acid, cyclohexane pentanoic acid, hexanedioic acid, cyclohexanebutanoic acid, and (4-methylphen
  • complexing perturbants which can form the supramolecular complexes described herein are within the scope of the present invention
  • other examples of complexing perturbants include, but are not limited to, 2-carboxymethyl-phenylalanine-leucine; 2-benzyl succinic acid, an actinonin, phenylsulfonyl aminophenyl-butyric acid,
  • Mimetics within the scope of the present invention are constructs which are structural and/or functional equivalents of an original entity.
  • Structural and chemically functional mimetics of the supramolecular complexes and the reversible transportable intermediate states of active agents are not necessarily peptidic, as non-peptidic mimetics can be prepared which have the appropriate chemical and/or structural properties.
  • preferred mimetics are peptides which have a different primary structure than the supramolecular complex or the intermediate state, but retain the same secondary and tertiary structure of the supramolecular complex or the intermediate state.
  • mimetics may have less bioactivity than a native state or intermediate state active agent or supra molecular complex, the mimetics may possess other important properties which may not be possessed by the native state such as, for example, further increased ability to be delivered orally.
  • protein structures are determined by the collective intra- and inter-molecular interactions of the constituent amino acids.
  • the first and fourth amino acid in the helix interact non-covalently with one another. This pattern repeats through the entire helix except for the first four and last four amino acids.
  • the side chains of amino acids can interact with one another.
  • the phenyl side chain of phenylaline would probably not be solvent exposed if that phenylalanine were found in a helix. If the interactions of that phenylalanine contributed to helix stability then substituting an alanine for a phenylalanine would disrupt the helix and change the conformation of a protein.
  • a mimetic could be created by first determining which amino acid side chains became solvent exposed and thus removed from contributing to stabilization of the native state such as by the technique of scanning mutagenesis. Mutants containing amino acid substitutions at those same sights could be created so that the substituted amino acids would render the protein conformation more intermediate-like that native-like. Confirmation that the appropriate structure had been synthesized could come from spectroscopy and other analytical methods.
  • Delivery compositions which include the supramolecular complex described above are typically formulated by mixing an effective amount of perturbant appropriate for the chosen route of delivery i.e, subcutaneous, nasal, or sublingual, with the active agent.
  • the components can be prepared well prior to administration or can be mixed just prior to administration.
  • the delivery compositions of the present invention may also include one or more enzyme inhibitors.
  • enzyme inhibitors include, but are not limited to, compounds such as actinonin or epiactinonin and derivatives thereof. These compounds have the formulas below:
  • R 31 is sulfoxymethyl or carboxyl or a substituted carboxy group selected from carboxamide, hydroxyaminocarbonyl and alkoxycarbonyl groups; and R 32 is hydroxyl, alkoxy, hydroxyamino or sulfoxyamino group.
  • Other enzyme inhibitors include, but are not limited to, aprotinin (Trasylol) and Bowman-Birk inhibitor.
  • the stabilizing additives may be incorporated into a carrier solution of the supramolecular complex. With some drugs, the presence of such additives promotes the stability and dispersibility of the agent in solution.
  • the stabilizing 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, methyl cellulose, polyethylene glycol, carboxylic acids and salts thereof, and polylysine.
  • the preferred stabilizing additives are gum acacia, gelatin and methyl cellulose.
  • the amount of active agent is an amount effective to accomplish the purpose of the particular active agent.
  • the amount in the composition typically is a pharmacologically or biologically effective amount. However, the amount can be less than a pharmacologically or biologically effective amount when the composition is used in a dosage unit form, such as a capsule, a tablet or a liquid, because the dosage unit form may contain a multiplicity of carrier/biologically or chemically active agent compositions or may contain a divided pharmacologically or biologically effective amount.
  • the total effective amounts can then be administered in cumulative units containing, in total, pharmacologically or biologically or chemically active amounts of biologically or pharmacologically active agent.
  • the total amount of active agent, and particularly biologically or chemically active agent, to be used can be determined by those skilled in the art. However, it has surprisingly been found that with some biologically or chemically active agents, the use of the presently disclosed carriers provides extremely efficient delivery, particularly in, intranasal, sublingual, or subcutaneous systems. Therefore, lower amounts of biologically or chemically active agent than those used in prior dosage unit forms or delivery systems can be administered to the subject, while still achieving the same blood levels and therapeutic effects.
  • the amount of perturbant used is a delivery effective amount.
  • compositions of the present invention may be formulated into dosage units by the addition of one or more excipient(s), diluent(s), disintegrant(s), lubricant(s), plasticizer(s), colorant(s), or dosing vehicle(s).
  • Preferred dosage unit forms are oral dosage unit forms. Most preferred dosage unit forms include, but are not limited to, tablets, capsules, or liquids.
  • the dosage unit forms can include biologically, pharmacologically, or therapeutically effective amounts of the active agent or can include less than such an amount if multiple dosage unit forms are to be used to administer a total dosage of the active agent. Dosage unit forms are prepared by methods conventional in the art.
  • the subject invention is useful for administering biologically active agents to any animals such as birds; mammals, such as primates and particularly humans; and insects.
  • the systems are particularly advantageous for delivering chemical or biologically active agents which would otherwise be destroyed or rendered less effective by conditions encountered before the active agent in the native state reaches its target zone (i.e. the area to which the active agent to be delivered) and by conditions within the body of the animal to which they are administered.
  • the present invention is useful in subcutaneously, intranasally, or sublingually administering active agents.
  • Lane 1 High molecular weight marker (Bio-Rad)—1:20 dilution w/dH 2 O—(5 ⁇ l—>100 ⁇ l).
  • Lane 2 ⁇ -interferon A (647 ⁇ g/mL) control 5 ⁇ l+5 ⁇ l Bromophenol Blue (BPB)—(1.29 ⁇ g loaded).
  • Example 2 The method of Example 1 was followed substituting the thermal condensation product of glutamic acid, aspartic acid, tyrosine, and phenyl-alanine (Glu-Asp-Tye-Phe) that was fractionated through a 3000 molecular weight cut-off filter for the perturbant.
  • Lane 1 High Molecular Weight marker (Bio-Rad).
  • Lane 2 ⁇ -interferon (647 ⁇ g/mL) ⁇ 5 ⁇ l+5 ⁇ l BPB control.
  • Examples 1 and 1A illustrate that ⁇ -interferon alone (lane 2 in FIGS. 1 and 2) banded at the appropriate molecular weight (approximately 19,000 Daltons). As the amount of perturbant added is increased in each subsequent lane relative to a fixed concentration of ⁇ -interferon, the ⁇ -interferon migrates to a lower, rather than a higher, molecular weight. The change seen with the perturbant of Example 1 is more pronounced than that seen with the perturbant of Example 1A. This indicates that the ⁇ -interferon structure is changing due to the two different perturbants, because if structure were not changing, there would be a shift towards higher molecular weight as perturbant complexes with the active agent.
  • a dosing composition of the perturbant of Example 1 at 2.4 mM and sCt at 0.3 mM was prepared, and isothermal titration calorimetry was performed at pH 6.5 and 4.5.
  • the buffer at pH 6.5 was 30 mM Hepes-30 mM NaCl, and the buffer at pH 4.5, was 30 mM sodium acetate-30 mM NaCl.
  • Example 2 The method of Example 2 was followed substituting the perturbant of Example 1A. Results were validated in experiments where perturbant was placed in the dropping syringe, and equivalent increments were added to pH 4.5 buffer (no sct).
  • a stock solution of 9.1 mg/mL of ⁇ interferon (Schering Plough Corp.) in 20 mM sodium phosphate buffer at pH 7.2 was prepared. Samples were prepared by diluting the ⁇ -interferon with the sodium phosphate buffer and 10 M guanidine hydrochloride (GuHCl) (Sigma Chemical Co.—St. Louis, Mo.) stock solution to 200 ⁇ g/mL concentration of ⁇ -interferon at various concentrations of GuHCl. Diluted samples were allowed to come to equilibrium by incubation for approximately 30 minutes at room temperature prior to measurement.
  • ⁇ interferon Schering Plough Corp.
  • Fluorescence measurements were made at 25° C. using a Hitachi F-4500. Protein tryptophan fluorescence was observed at an excitation wavelength of 298 nm and an emission wavelength of 343 nm. ANS (1-anilinonapthalene-8-sulfonate) fluorescence was observed at an excitation wavelength of 355 nm and an emission wavelength of 530 nm. For all fluorescence measurements, a 5 nm spectral bandpass was chosen for both excitation and emission.
  • GuHCl 5M stock solution was prepared using 20 mM sodium phosphate, pH 7.2 buffer. After dilution, the pH of the stock was checked and adjusted by concentrated HCl. To determine the concentration of final solution the refractive index referenced in Methods in Enzymology, Vol. 6, page 43 by Yasuhiko Nozaki was used.
  • ⁇ -interferon stock (9.1 mg/mL) was mixed with sufficient amounts of GuHCl to yield the concentrations of Table 1A below: TABLE 1A ⁇ -Interferon/GuHCI Solutions GuHCI (M) ⁇ -IFN (mg/mL) 0.5 0.60 1.0 0.53 1.5 0.60 2.0 0.50 3.0 0.60 4.0 0.50
  • DSC Differential scanning calorimetry
  • a stock solution of 9.1 mg/mL ⁇ -interferon in 20 mM sodium phosphate buffer at pH 7.2 was prepared. Samples were prepared by diluting the ⁇ -interferon to a concentration of 200 ug/mL into solution buffered at various pH values using the following buffers: Glycine at pH 2 and 12, sodium phosphate at pH 3, 4, 5, 7, and boric acid at pH 8. These buffers were prepared as described in the Practical Handbook of Biochemistry and Molecular Biology, Edited by Gerald D. Fasman, 1990. Diluted samples were allowed to come to equilibrium by incubation for approximately 30 minutes at room temperature prior to measurement.
  • a stock solution was prepared by dissolving 2 mg of insulin in 1 mL of deionized water.
  • 1-anilinonaphthalene-8-sulphonate (ANS) stock solution was prepared by dissolving 10 mg in 10 mL of deionized water.
  • Samples were prepared by diluting the insulin to a concentration of 200 ug/mL into solution buffered at various pH values using the following buffers: Glycine at pH 2 and 12, sodium phosphate at pH 3, 4, 5, 7, and boric acid at pH 8. These buffers were prepared as described in the Practical Handbook of Biochemistry and Molecular Biology, Edited by Gerald D. Fasman, 1990. The final ANS concentration was 90 ug/mL. Diluted samples were allowed to come to equilibrium by incubation for approx. 30 minutes at room temperature prior to measurement.
  • Circular dichroism spectra of ⁇ -interferon were generated at pH 7.2. The pH of the solution was then readjusted to pH 2, and the sample was rescanned. The sample solution was then readjusted to 7.2 and rescanned.
  • the secondary structure content was estimated with several fitting programs, each of which decomposes the CD curve into four major structural components: ⁇ -helix, ⁇ -sheet, turns, and random coil. Two of those programs were provided with the CD instrument as an analysis facility. The first program uses seven reference proteins: Myoglobin, Lysozyme, Papain, Cytochrome C, Hemoglobin, Ribonuclease A and Chymotrypsin. The second uses Yang.REF reference file.
  • a third program, CCAFAST uses the Convex Constraint Algorithm and is described in “Analysis of Circular Dichroism Spectrum of Proteins Using the Convex Constraint Algorithm: A Practical Guide”. (A. Perczel, K. Park and G. D. Fasman (1992) Anal. Biochem. 203: 83-93).
  • DSC was performed with 6 mg/mL insulin (0.83 mM assuming a molecular weight of 6,000) in 50 mM phosphate buffer, pH 7.5. Each subsequent thermogram was corrected by background subtraction of a 0.6M guanidine-phosphate buffer solution.
  • Insulin was freshly prepared as a concentrated stock solution in 50 mM phosphate buffer, pH 7.5, and an appropriate aliquot was diluted in buffer, filtered though a 2 micron PTFE filter, and degassed for at least 20 minutes.
  • the reference cell contained degassed buffer.
  • Example 9 The method of Example 9 was followed substituting 5 mg/mL recombinant human growth hormone (rhGh) (225 ⁇ M based on M,22,128 of HGH) in 50 mM phosphate buffer, pH 7.5 containing either 0.5 or 1.0M NaCl, for the insulin. The thermograms were corrected by subtraction of a 0.5M NaCl-phosphate buffer blank.
  • rhGh human growth hormone
  • ⁇ -interferon stock (9.1 mg/mL) was diluted with buffer to a concentration of 0.6 mg/mL. The sample was dialyzed overnight in buffer (volume ratio of ⁇ -interferon to buffer was 1:4000). Since there was no extinction coefficient provided, concentration of the sample used was determined by comparison of absorption spectra of the sample before and after dialysis. For each particular pH, the absorbance of the nondialyzed ⁇ -interferon of known concentration was measured at 280 nm. Then after dialysis, absorbance was read again to account for the protein loss, dilution, etc. Buffer conditions and ⁇ -interferon concentrations were:
  • pH 4.1 Buffer—20 mM NaAc.
  • IFN 0.53 mg/mL;
  • DSC scans were performed according to the procedure of Example 9. Although clear, transparent solutions of ⁇ -interferon were obtained for every pH at room temperature, there were noticeable signs of precipitation at pH 5.0 and 6.0 after the temperature scans.
  • GuHCL/ ⁇ -interferon samples were prepared according to the method of Example 4. DSC scans were performed according to the procedure of Example 9.
  • Examples 3-14 illustrate that ionic strength, guanidine hydrochloride concentration, and pH result in changes in the Tm of active agents, indicating a change in conformation. This was confirmed by fluorescence spectroscopy. The reversible intermediate conformational states can be used as templates to prepare mimetics.
  • An intermediate conformational state of ⁇ -interferon is determined.
  • a peptide mimetic having the secondary and tertiary structure of the intermediate state is prepared.
  • Example 15 The method of Example 15 is followed substituting an insulin for the ⁇ -interferon.
  • Example 15 The method of Example 15 is followed substituting recombinant human growth hormone for the ⁇ -interferon.
  • a stock solution of 9.1 mg/mL ⁇ -interferon in 20 mM sodium phosphate buffer at pH 7.2 was prepared.
  • a stock solution of perturbant was prepared by dissolving 800 mg of perturbant (L-arginine acylated with cyclohexanoyl chloride) in 2 mL of 20 mM Sodium Phosphate buffer (pH7).
  • Samples were prepared by diluting the ⁇ -interferon with the sodium phosphate buffer and perturbant stock solution at various perturbant concentrations. Diluted samples were allowed to come to equilibrium by incubation for approximately 30 minutes at room temperature prior to measurement.
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant was weighed out to make perturbant stock solutions. ⁇ -interferon stock was diluted in the buffer. ⁇ -interferon solution was not dialyzed prior to experiments for the purpose of having the same active concentration for the whole set.
  • DSC thermograms were generated with ⁇ -interferon at a concentration of 0.64 mg/ml and a perturbant (phenylsulfonyl-para-aminobenzoic acid purified to >98% (as determined by reverse phase chromatography prior to generation of the spectra)) at perturbant concentrations of 5, 10, 25 and 100 mg/ml.
  • DSC was conducted on a DASM-4 differential scanning calorimeter interfaced to an IBM PC for automatic collection of the data. The scanning rate was 60° C./h.
  • Example 19 The method of Example 19 was followed substituting ⁇ -interferon without perturbant. Results are illustrated in Table 9 below and FIG. 10. TABLE 9 ⁇ -Interferon + Perturbant DSC Perturbant - mg/ml Tm ° C. ⁇ H cal/mol 0 67.12 72562 5 64.37 60151 10 62.3 53161 25 58.15 35393 100 46.18 5439.3
  • the dialyzed sample had essentially the same Tm and the same area under the Cp vs. Tm curve as it did prior to addition of the perturbant. This indicated that not only was the perturbant able to induce conformational changes in the protein but that this process was reversible. Dilution was enough of a driving force to effect disengagement of the perturbant from the active agent.
  • Example 4 The method of Example 4 was followed, substituting the perturbant of Example 19 for the GuHCl.
  • Example 19 The method of Example 19 was followed substituting cyclohexanoyl phenylglycine for the perturbant.
  • Example 4 The method of Example 4 was followed substituting the perturbant of Example 30 for the GuHCl.
  • the ⁇ H v. Tm plot indicates the existence of an equilibrium intermediate conformation of ⁇ -interferon that is stable at below 5 and 25 mg/ml of added cyclohexanoyl phenylglycine perturbant.
  • Isothermal titration calorimetry of perturbant complexing with ⁇ -interferon was performed at 25° C. at two different pH's.
  • the buffers used were 20 mM NaPhosphate for pH 7.2 and 20 mM NaAc for pH 3.0.
  • ⁇ -interferon solution was dialyzed before the experiment to reach the appropriate pH. Dry perturbants were weighed and diluted in dialysate.
  • ITC was conducted on a MicroCal OMEGA titration calorimeter (MicroCal Inc.—Northampton, Mass.). Data points were collected every 2 seconds, without subsequent filtering. ⁇ -interferon solution placed in 1.3625 mL cell was titrated using a 250 ⁇ L syringe filled with concentrated perturbant solution. A certain amount of titrant was injected every 3-5 minutes for up to 55 injections.
  • x-axis units are concentration of carrier in mM.
  • y-axis units represent heat/injection expressed in calories.
  • Example 24 and Comparative Example 24* illustrate that ⁇ -interferon has a positive enthalpy and a binding constant (K d ⁇ 10 ⁇ 3 M).
  • Example 24 The method of Example 24 was followed substituting the perturbant of Example 19 for the perturbant of Example 22.
  • x-axis units are concentration of carrier in mM.
  • y-axis units represent heat/injection expressed in calories.
  • Examples 24 and 25 indicate that the stronger the perturbant complexes with the active agent and the more thermodynamically stable the intermediate state of the active agent, the greater the bioavailability of the active agent.
  • FIGS. 17 and 18 illustrate that conformational changes in ⁇ -interferon are more readily produced by benzoyl para-amino phenylbutyric acid than by the perturbants of Examples 22 and 19, and that such changes are more readily produced by the perturbant of Example 22 than by the perturbant of Example 19.
  • x-axis represents concentration of carrier in mM.
  • y-axis represents heat/injection expressed in calories.
  • the apparent dissociation constant for the perturbant of Example 26 is greater than that for the perturbant of Example 30 (10 ⁇ 4 M) at pH 7.
  • benzoyl para-amino phenylbutyric acid complexes more strongly to ⁇ -interferon and induces the native state-reversible intermediate conformational state at lower concentrations of perturbant.
  • the sum total of the heat evolved was plotted against the total perturbant concentration to produce the isotherm from which the association constant (K A , M), enthalpy change ( ⁇ H, kcal/mol), entropy change ( ⁇ S (eu), and N, and the stoichiometry of perturbant molecules complexed per equivalent of complexed supramolecular complex, were determined by curve-fitting the binding isotherm against the binding equation described for perturbant complexing in a supramolecular complex possessing one set of independent perturbant complexing sites.
  • the data were deconvoluted using the nonlinear least squares algorithm supplied in the software of the manufacturer.
  • the assay for pancreatin activity was prepared as follows: 0.1 mL of a stock solution of ⁇ -interferon (9.1 mg/mL, 20 mM NaH 2 PO 4 , pH 7.2) (Schering-Plough Corp.) was added to 2.5 mL of either phenylsulfonyl-para-aminobenzoic acid perturbant (46) or cyclohexanoyl phenylglycine perturbant (47) (200 mg/mL) in 5 mM KH 2 PO 4 , pH 7.0. Incubation was carried out at 37° C.
  • Examples 31 and 32 illustrate that both supramolecular complexes resisted enzymatic degradation. However, in additional testing no correlation was shown between the enzyme inhibitors potency and the ability to deliver drug.
  • the mobil phase was 67 mM phosphate buffer, pH7.4.
  • the flow rate was 1.5 mL/min isocratic.
  • Sample detection was done with a Perkin Elmer refractive index detector.
  • K′ (Ret. time Carrier/Ret. time Water) ⁇ 1
  • DSC experiments were carried out according to the procedure of Example 19, with 1.0 mg/ml dipalmitoylphosphatidylcholine (DPPC) mixed with perturbants XI, L, LII, LIII, and LIV.
  • the concentrations of the perturbant were varied from 0 to 20 mg/ml.
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant L was weighed out to make perturbant stock solutions. DPPC stock solution was prepared in the buffer.
  • DSC thermograms were generated with DPPC at a concentration of 1.0 mg/ml.
  • the perturbant concentrations used were 0, 5, 10, and 20 mg/ml.
  • the DSC was performed as described in Example 19.
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant L was weighed out to make perturbant stock solutions.
  • DPPC stock solution was prepared in the buffer.
  • rhGH solution was prepared as described in Example 10A.
  • DSC thermograms were generated with DPPC at a concentration of 1.0 mg/ml. Samples having DPPC alone; DPPC with 10 mg/ml of perturbant; DPPC with 0.3 mg/ml of rhGH; and DPPC, 10 mg/ml of perturbant and 0.3 mg/ml of rhGH were prepared and analyzed. The DSC was performed as described in Example 19.
  • DSC thermograms were generated with DPPC at a concentration of 1.0 mg/ml. Samples having DPPC alone; DPPC with 5 mg/ml of perturbant; DPPC with 0.3 mg/ml of rhGH; and DPPC, 5 mg/ml of perturbant and 0.3 mg/ml of rhGH were prepared and analyzed. The DSC was performed as described in Example 19.
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant XI was weighed out to make perturbant stock solutions.
  • DPPC stock solution was prepared in the buffer.
  • rhGH solution was prepared as described in Example 10A.
  • DSC thermograms were generated with DPPC at a concentration of 1.0 mg/ml. Samples having DPPC alone; DPPC with 2 mg/ml of perturbant; DPPC with 0.3 mg/ml of rhGH; and DPPC, 2 mg/ml of perturbant and 0.3 mg/ml of rhGH were prepared and analyzed. The DSC was performed as described in Example 19.
  • Dosing compositions were prepared by mixing the modified amino acids and recombinant human growth hormone (rhGH) as listed in Table 16 below in a phosphate buffer solution at a pH of about 7-8.
  • Rats were administered the dosing composition by sublingual or intranasal administration. Delivery was evaluated by using an ELISA assay for rhGH from Medix Biotech, Inc. For intracolonic administration, a sample was prepared and dosed to fasted rats at 25 mg/kg of carrier in a buffered solution containing propylene glycol (0-50%) and 1 mg/kg rhGH.
  • rhGH (6 mg/ml) was administered by oral gavage to a rat, and delivery was evaluated according to the procedure of Example 41A.
  • Results are illustrated in Table 16 below.
  • TABLE 16 In Vivo Delivery of rhGH Carrier Drug Method of Mean Peak Serum Dose Dose Admin- Levels of rhGH
  • the delivery agents were reconstituted with distilled water and adjusted to pH 7.2-8.0 with either aqueous hydrochloric acid or aqueous sodium hydroxide.
  • a stock solution of rhGH was prepared by mixing rhGH, D-mannitol and glycine and dissolving this mixture in 2% glycerol/water. The stock solution was then added to the delivery agent solution.
  • Several delivery agent to active agent ratios were studied.
  • Serum rhGH concentrations were quantified by an rhGH enzyme immunoassay test kit. The results are given in Table 17 and FIGS. 28 and 29.
  • FIG. 29 the circles represent the response following SL dosing of an aqueous solution of compound CXXIII-H and rhGH.
  • the squares represent the response following IN dosing of an aqueous solution of compound CXXIII-H and rhGH.
  • the triangles represent the response following IC dosing of an aqueous solution of compound CXXIII-H and rhGH.
  • the dose of compound CXXIII-H was 25 mg/kg and the dose of rhGH was 1 mg/kg.
  • rhGH (1 mg/kg) was administered by oral gavage to a rat, and delivery was evaluated according to the procedure of Example 43.
  • the delivery agents were reconstituted with distilled water and adjusted to pH 7.2-8.0 with either aqueous hydrochloric acid or aqueous sodium hydroxide.
  • a stock solution of sCT was prepared by dissolving sCT in citric acid (0.085N). The stock solution was then added to the delivery agent solution.
  • Several different delivery agent to active agent ratios were studied.
  • Serum calcium concentrations were quantified by the o-cresolphthalein complexone method (Sigma) using a UV/VIS spectrophotometer (Perkin Elmer). The results are given in Table 5.
  • Salmon calcitonin was administered by oral gavage to rats, and delivery was evaluated according to the Procedure of Example 55. The results are given in Table 18 below. TABLE 18 Delivery Agent Enhancement of Salmon Calcitonin (sCT, dosed at 0.2 ⁇ g/kg) Bioavailability Administered by Subcutaneous Administration. Delivery Agent Dose Percent Decrease in Example Deliver Ageny ( ⁇ g/kg) Serum Calcium 55 CXXIII-H 2 17 ⁇ 3 55A None 0 17 ⁇ 2 56 CXXIII-H 20 25 ⁇ 4 57 CXXIII-H 200 25 ⁇ 5 58 CXXIII-H 2000 26 ⁇ 5 59 CXI-G 20 21 ⁇ 4 60 CXIV-G 20 20 ⁇ 3

Abstract

Methods for transporting a biologically active agent across a cellular membrane or a lipid bilayer. A first method includes the steps of:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to the native state and which is conformationally between the native and denatured states;
(b) exposing the biologically active agent to a complexing perturbant to reversibly transform the biologically active agent to the intermediate state and to form a transportable supramolecular complex; and
(c) exposing the membrane or bilayer to the supramolecular complex, to transport the biologically active agent across the membrane or bilayer. The perturbant has a molecular weight between about 150 and about 600 daltons, and contains at least one hydrophilic moiety and at least one hydrophobic moiety. The supramolecular complex comprises the perturbant non-covalently bound or complexed with the biologically active agent. In the present invention, the biologically active agent does not form a microsphere after interacting with the perturbant. A method for preparing an orally administrable biologically active agent comprising steps (a) and (b) above is also provided as are oral delivery compositions.
Additionally, mimetics and methods for preparing mimetics are contemplated.

Description

  • This application is a continuation-in-part of: [0001]
  • (a) U.S. Ser. No. 08/763,183, filed Dec. 10, 1996, which is a continuation-in-part of U.S. Ser. No. 08/328,932, filed Oct. 25, 1994; [0002]
  • (b) U.S. Ser. No. 08/051,019, filed Apr. 22, 1993, now U.S. Pat. No. 5,451,410; [0003]
  • (c) U.S. Ser. No. 08/168,776, filed Dec. 16, 1993, now U.S. Pat. No. 5,447,728, which is a continuation-in-part of U.S. Ser. No. 08/051,019, filed Apr. 22, 1993, now U.S. Pat. No. 5,451,410, and of U.S. Ser. No. 08/143,571, filed Oct. 26, 1993, which is a continuation-in-part of U.S. Ser. No. 08/076,803, filed Jun. 14, 1993, which is a continuation-in-part of U.S. Ser. No. 07/920,346, filed Jul. 27, 1992, now U.S. Pat. No. 5,443,841, which is a continuation-in-part of U.S. Ser. No. 07/898,909, filed Jun. 15, 1992; [0004]
  • (d) PCT Serial No. PCT/US94/04560, filed Apr. 22, 1994, which is a continuation-in-part of U.S. Ser. No. 08/051,019, filed Apr. 22, 1993, now U.S. Pat. No. 5,451,410, and of U.S. Ser. No. 08/205,511, filed on Mar. 2, 1994; [0005]
  • (e) U.S. Ser. No. 08/231,622, filed Apr. 22, 1994; [0006]
  • (f) U.S. Ser. No. 08/205,511, filed Mar. 2, 1994; [0007]
  • (g) U.S. Ser. No. 08/231,623, filed Apr. 22, 1994, now U.S. Pat. No. 5,541,155; [0008]
  • (h) U.S. Ser. No. 08/315,200, filed Sep. 29, 1994; [0009]
  • (i) U.S. Ser. No. 08/316,404, filed Sep. 30, 1994; and [0010]
  • (j) U.S. Ser. No. 08/820,644, filed Mar. 18, 1997, which is a conversoin of U.S. provisional patent application Serial No. 60/017,902, filed Mar. 29, 1996.[0011]
  • FIELD OF THE INVENTION
  • The present invention relates to methods and compositions for transporting active agents, and particularly biologically active agents, across cell membranes or lipid bilayers. These methods and compositions facilitate the delivery of an active agent to a target, such as the delivery of a pharmaceutical agent through an adverse environment to a particular location of the body. [0012]
  • BACKGROUND OF THE INVENTION
  • Conventional means for delivering active agents to their intended targets, e.g. human organs, tumor cites, etc., are often severely limited by the presence of biological, chemical, and physical barriers. Typically, these barriers are imposed by the environment through which delivery must take place, the environment of the target for delivery, or the target itself. [0013]
  • Biologically active agents are particularly vulnerable to such barriers. In the delivery to animals of such agents, including, but not limited to pharmacological and therapeutical agents, barriers are impeded by the body. Subcutaneous, nasal or sublingual delivery to the circulatory system for many biologically active agents would be the route of choice for administration to animals if not for physical barriers such as the skin, lipid bi-layers, and various organ membranes that are relatively impermeable to certain biologically active agents, but one or more of which must be traversed before an agent delivered via these routes can reach the circulatory system. Additionally, delivery such as, for example, sublingual delivery may be impeded by chemical barriers such as the varying pH in the gastrointestinal (GI) tract and the presence of powerful digestive enzymes. [0014]
  • While in many cases different methods of administration of these compounds would be preferable. Many of these agents cannot be delivered by these routes to the target at which the active agent renders its intended biological effect. [0015]
  • Typically, the initial focus of drug design is on the physiochemical properties of pharmaceutical compounds and particularly their therapeutic function. The secondary design focus is on the need to deliver the active agent to its biological target(s). This is particularly true for drugs and other biologically active agents that are designed for oral administration to humans and other animals. However, thousands of therapeutic compounds are discarded because no delivery systems are available to ensure that therapeutic titers of the compounds will reach the appropriate anatomical location or compartment(s) after administration and particularly oral administration. Furthermore, many existing therapeutic agents are underutilized for their approved indications because of constraints on their mode(s) of administration. Additionally, many therapeutic agents could be effective for additional clinical indications beyond those for which they are already employed if there existed a practical methodology to deliver them in appropriate quantities to the appropriate biological targets. [0016]
  • Although nature has achieved successful inter- and intra-cellular transport of active agents such as proteins, this success has not been translated to drug design. In nature, the transportable conformation of an active agent such as a protein is different than the conformation of the protein in its native state. In addition, natural transport systems often effect a return to the native state of the protein subsequent to transport. When proteins are synthesized by ribosomes, they are shuttled to the appropriate cellular organelle by a variety of mechanisms e.g. signal peptides and/or chaperoning. Gething, M-J., Sambrook, J., [0017] Nature, 355, 1992, 33-45. One of the many functions of either the signal peptides or the chaperonins is to prevent premature folding of the protein into the native state. The native state is usually described as the 3-dimensional state with the lowest free energy. By maintaining the protein in a partially unfolded state, the signal peptides or the chaperonins facilitate the protein's ability to cross various cellular membranes until the protein reaches the appropriate organelle. The chaperonin then separates from the protein or the signal peptide is cleaved from the protein, allowing the protein to fold to the native state. It is well known that the ability of the protein to transit cellular membranes is at least partly a consequence of being in a partially unfolded state.
  • Current concepts of protein folding suggest that there are a number of discrete conformations in the transition from the native state to the fully denatured state. Baker, D., Agard, D. A., [0018] Biochemistry, 33, 1994, 7505-7509. The framework model of protein folding suggests that in the initial early stages of folding the domains of the protein that are the secondary structure units will form followed by the final folding into the native state. Kim, P. S., Baldwin, R. L., Annu. Rev. Biochem., 59, 1990, 631-660. In addition to these kinetic intermediates, equilibrium intermediates appear to be significant for a number of cellular functions. Bychkova, V. E., Berni, R., et al, Biochemistry, 31, 1992, 7566-7571, and Sinev, M. A., Razgulyaev, O. I., et al, Eur. J. Biochem., 1989, 180, 61-66. Available data on chaperonins indicate that they function, in part, by keeping proteins in a conformation that is not the native state. In addition, it has been demonstrated that proteins in partially unfolded states are able to pass through membranes, whereas the native state, especially of large globular proteins, penetrates membranes poorly, if at all. Haynie, D. T., Freire, E., Proteins:Structure, Function and Genetics, 16, 1993, 115-140.
  • Similarly, some ligands such as insulin which are unable to undergo conformational changes associated with the equilibrium intermediates described above, lose their functionality. Hua, Q. X., Ladbury, J. E., Weiss, M. A., [0019] Biochemistry, 1993, 32, 1433-1442; Remington, S., Wiegand, G., Huber, R., 1982, 158, 111-152; Hua, Q. X., Shoelson, S. E., Kochoyan, M. Weiss, M. A., Nature, 1991, 354, 238-241.
  • Studies with diphtheria toxin and cholera toxin indicate that after diphtheria toxin binds to its cellular receptor, it is endocytosed, and while in this endocytic vesicle, it is exposed to an acidic pH environment. The acidic pH induces a structural change in the toxin molecule which provides the driving force for membrane insertion and translocation to the cytosol. See, Ramsay, G., Freire, E. [0020] Biochemistry, 1990, 29, 8677-8683 and Schon, A., Freire, E., Biochemistry, 1989, 28, 5019-5024. Similarly, cholera toxin undergoes a conformational change subsequent to endocytosis which allows the molecule to penetrate the nuclear membrane. See also, Morin, P. E., Diggs, D., Freire, E., Biochemistry, 1990, 29, 781-788.
  • Earlier designed delivery systems have used either an indirect or a direct approach to delivery. The indirect approach seeks to protect the drug from a hostile environment. Examples are enteric coatings, liposomes, microspheres, microcapsules. See, colloidal drug delivery systems, 1994, ed. Jorg Freuter, Marcel Dekker, Inc.; U.S. Pat. No. 4,239,754; Patel et al. (1976), [0021] FEBS Letters, Vol. 62, pg. 60; and Hashimoto et al. (1979), Endocrinology Japan, Vol. 26, pg. 337. All of these approaches are indirect in that their design rationale is not directed to the drug, but rather is directed to protecting against the environment through which the drug must pass enroute to the target at which it will exert its biological activity, i.e. to prevent the hostile environment from contacting and destroying the drug.
  • The direct approach is based upon forming covalent linkages with the drug and a modifier, such as the creation of a prodrug. Balant, L. P., Doelker, E., Buri, P., [0022] Eur. J. Drug Metab. And Pharmacokinetics, 1990, 15(2), 143-153. The linkage is usually designed to be broken under defined circumstances, e.g. pH changes or exposure to specific enzymes. The covalent linkage of the drug to a modifier essentially creates a new molecule with new properties such as an altered log P value and/or as well as a new spatial configuration. The new molecule has different solubility properties and is less susceptible to enzymatic digestion. An example of this type of method is the covalent linkage of polyethylene glycol to proteins. Abuchowski, A., Van Es, T., Palczuk, N. C., Davis, F. F., J. Biol. Chem. 1977, 252, 3578.
  • Broad spectrum use of prior delivery systems has been precluded, however, because: (1) the systems require toxic amounts of adjuvants or inhibitors; (2) suitable low molecular weight cargos, i.e. active agents, are not available; (3) the systems exhibit poor stability and inadequate shelf life; (4) the systems are difficult to manufacture; (5) the systems fail to protect the active agent (cargo); (6) the systems adversely alter the active agent; or (7) the systems fail to allow or promote absorption of the active agent. [0023]
  • There is still a need in the art for simple, inexpensive delivery systems which are easily prepared and which can deliver a broad range of active agents to their intended targets, especially in the case of pharmaceutical agents that are to be administered via the oral route. [0024]
  • SUMMARY OF THE INVENTION
  • The present invention discloses methods for administering, by either the subcutaneous, nasal, or sublingual routes, a biologically active agent to a subject in need of such agent. A first method includes the steps of: [0025]
  • (a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to the native state and which is conformationally between the native and denatured states; [0026]
  • (b) exposing the biologically active agent to a complexing perturbant to reversibly transform the biologically active agent to the intermediate state and to form a transportable supramolecular complex wherein the perturbant is in an amount effective for subcutaneous, nasal or sublingual delivery of the biologically active agent; and [0027]
  • (c) either subcutaneously, nasally, or sublingually administering the supramolecular complex to the subject. [0028]
  • The perturbant has a molecular weight between about 150 and about 600 daltons, and contains at least one hydrophilic moiety and at least one hydrophobic moiety. The supramolecular complex comprises the perturbant non-covalently bound or complexed with the biologically active agent. In the present invention, the biologically active agent does not form a microsphere after interacting with the perturbant. [0029]
  • Also contemplated is a method for preparing subcutaneously, nasally, or sublingually administrable biologically active agent comprising steps (a) and (b) above. [0030]
  • In alternate embodiments, subcutaneous, nasal, or sublingual delivery compositions are provided. The compositions comprise a supramolecular complex including: [0031]
  • (a) a biologically active agent in an intermediate conformational state which is reversible to the native state, non-covalently complexed with [0032]
  • (b) a complexing perturbant having a molecular weight ranging from about 150 to about 600 and having at least one hydrophilic moiety and at least one hydrophobic moiety wherein the perturbant is in an amount effective for subcutaneous, nasal, or sublingual delivery of the biologically active agent; and [0033]
  • wherein the intermediate state is conformationally between the native conformation state and denatured conformation state of the biologically active agent and the composition is not a microsphere. [0034]
  • Further contemplated is a method for preparing an agent which is a mimetic which is capable of being administered, by the subcutaneous, nasal, or sublingual route, to a subject in need of such agent. A biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to the native state and which is conformationally between the native state and the denatured state, is exposed to a complexing perturbant in an amount effective for subcutaneous, nasal, or sublingual delivery of such an agent to reversibly transform the biologically active agent to the intermediate conformational state and to form a transportable supramolecular complex. The perturbant has a molecular weight between about 150 and about 600 daltons and at least one hydrophilic moiety and one hydrophobic moiety. The supramolecular complex comprises the perturbant non-covalently complexed with the biologically active agent, and the biologically active agent does not form a microsphere with the perturbant. A mimetic of the supramolecular complex is prepared. [0035]
  • Alternatively, a method for preparing an agent which is capable of being administered either by the subcutaneous, nasal, or sublingual routes, is provided. A biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to the native state and which is conformationally between the native and denatured states, is exposed to a perturbant to reversibly transform the biologically active agent to the intermediate state. The perturbant is in a respective amount effective for subcutaneous, nasal, or sublingual delivery of the agent. The agent, a mimetic of the intermediate state, is prepared. [0036]
  • The administration by these routes in the methods and compositions of the present invention results in delivery of the agent in bioavailable and bioactive form.[0037]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is illustration of a native gradient gel of α-interferon (IFN) and a modified amino acid complexing perturbant. [0038]
  • FIG. 2 is an illustration of a native gradient gel of α-interferon and a thermal condensate complexing perturbant. [0039]
  • FIG. 3 is a graphic illustration of guanidine hydrochloride (GuHCl) induced denaturation of α-interferon. [0040]
  • FIG. 4 is a graphic illustration of the concentration effect of GuHCl on α-interferon conformation. [0041]
  • FIG. 5 is a graphic illustration of the pH denaturation of α-interferon. [0042]
  • FIG. 6 is a graphic illustration of the pH denaturation of insulin. [0043]
  • FIGS. 7A and 7B are graphic illustrations of the reversibility of the circular dichroism spectrum of α-interferon. [0044]
  • FIG. 8 is a graphic illustration of the circular dichroism spectrum of α-interferon. [0045]
  • FIG. 9 is a graphic illustration of intrinsic tryptophan fluorescence of α-interferon and a complexing perturbant. [0046]
  • FIG. 10 is a graphic illustration of the differential scanning calorimetry of α-interferon and complexing perturbant. [0047]
  • FIGS. 11A and 11B are graphic illustrations of the reversibility of the transformation due to complexing perturbants. [0048]
  • FIG. 12 is a graphic illustration of the effect of complexing perturbant on α-interferon. [0049]
  • FIG. 13 is a graphic illustration of the concentration effect of complexing perturbant on α-interferon conformation. [0050]
  • FIG. 14 is a graphic illustration of the effect of complexing perturbant on α-interferon. [0051]
  • FIG. 15 is a graphic illustration of the Isothermal Titration Calorimetry of α-interferon and complexing perturbant. [0052]
  • FIG. 16 is a graphic illustration of the Isothermal Titration Calorimetry of α-interferon and complexing perturbant. [0053]
  • FIG. 17 is a graphic illustration of the effects of complexing perturbants on α-interferon. [0054]
  • FIG. 18 is a graphic illustration of the effect of the concentration of complexing perturbants on α-interferon. [0055]
  • FIG. 19 is a graphic illustration of the Isothermal Titration Calorimetry of α-interferon and complexing perturbant. [0056]
  • FIG. 20 is a graphic illustration of pancreatic inhibition assay with α-interferon and complexing perturbants. [0057]
  • FIG. 21 is a graphic illustration of the effect of DSC of heparin at pH 5.0. [0058]
  • FIG. 22 is a graphic illustration of the DSC of DPPC with perturbant compounds at several concentrations (units T[0059] M,° C. vs. concentration).
  • FIG. 23 is a graphic illustration of the concentration effect of complexing perturbant compound L on the DPPC conformation. [0060]
  • FIG. 24 is a graphic illustration of the concentration effect of complexing perturbant compound L on rhGH conformation. [0061]
  • FIG. 25 is a graphic illustration of the concentration effect of complexing perturbant compound LI on rhGH conformation. [0062]
  • FIG. 26 is a graphic illustration of the concentration effect of complexing perturbant compound XI on rhGH conformation. [0063]
  • FIG. 27 is a graphic illustration of the differential light scattering of perturbant compound L in a 10 mM phosphate buffer at pH 7.0. [0064]
  • FIG. 28 is a graphic illustration of the results of subcutaneous injection of rhGH composition in rats. [0065]
  • FIG. 29 is a graphic illustration of the results of Sublingual (SL), intranasal (IN), and intracolonic (IC) dosing of rhGH in rats. [0066]
  • FIG. 30 is a graphic illustration of the results of intracolonic dosing of heparin with compound XXXI carrier. [0067]
  • DETAILED DESCRIPTION OF THE INVENTION
  • Subcutaneous, sublingual, and intranasal coadministration of an active agent, such as recombinant human growth hormone (rhGH), and the delivery agents, and particularly proteins, described herein results in an increased bioavailability of the active agent compared to administration of the active agent alone. A similar result is obtained by coadministration of salmon calcitonin with the delivery agents, in rats. Data supporting these findings are presented in the examples. [0068]
  • The present methods effect active agent delivery by creating a reversibly non-covalently complexed supramolecule from the active agent and an amount of complexing perturbant appropriate for the route of delivery. As a result, the three-dimensional structure or conformation of the active agent is changed, but the chemical composition of the active agent molecule is not altered. This alteration in structure (but not composition) provides the active agent with the appropriate properties, such as, for example, solubility (log P) to cross or penetrate a physical or chemical barrier, membrane, or lipid bilayer, to resist enzymatic degradation and the like. Crossing refers to transport from one side of the cell membrane or lipid bilayer to the opposite side (i.e. from the outside or exterior to the inside or interior of a cell and/or visa versa), whether the cell membrane or lipid bilayer is actually penetrated or not. Additionally, the perturbed intermediate state of the active agent or the supramolecular complex itself can be used as a template for the preparation of mimetics which, accordingly, could be delivered to a target by the appropriate route, i.e., subcutaneous, nasal, or sublingual. After crossing the cell membrane or lipid bilayer, an active agent has biological activity and bioavailability, either by restoration to the native state or by retaining biological activity or bioavailability acquired in the intermediate state. The mimetic acts similarly after crossing the cell membrane or lipid bilayer. [0069]
  • Active Agents [0070]
  • The native conformational state of an active agent is typically described as the three dimensional state with the lowest free energy (ΔG). It is the state in which the active agent typically possesses the full complement of activity ascribed to the agent, such as the full complement of biological activity ascribed to a biologically active agent. [0071]
  • The denatured conformational state is the state in which the active agent has no secondary or tertiary structure. [0072]
  • Intermediate conformational states exist between the native and denatured states. A particular active agent may have one or more intermediate states. The intermediate state achieved by the present invention is structurally and energetically distinct from both the native and denatured states. Active agents useful in the present invention must be transformable from their native conformational state to an intermediate conformational state which can be administered by the route of choice and back to their native state, i.e. reversibly transformable, so that when the active agent reaches its target, such as when a delivered drug reaches the circulatory system, the active agent retains, regains, or acquires a biologically, pharmacologically, or therapeutically significant complement of its desired biological activity. Preferably the ΔG of the intermediate state ranges from about −20 Kcal/mole to about 20 Kcal/mole, and most preferably, it ranges from about −10 Kcal/mole to about 10 Kcal/mole, all relative to the native state. [0073]
  • For example in the case of a protein, the intermediate state has significant secondary structure, significant compactness due to the presence of a sizable hydrophobic core, and a tertiary structure reminiscent of the native fold but without necessarily exhibiting the packing of the native state. The difference in free energy (ΔG) between the intermediate state and the native state is relatively small. Hence, the equilibrium constant between the native and the transportable, reversible intermediate state(s) is close to unity (depending upon experimental conditions). Intermediate states can be confirmed by, for example, differential scanning calorimetry (DSC), isothermal titration calorimetry (ITC), native gradient gels, NMR, fluorescence, and the like. [0074]
  • Without being bound by any theory, applicants believe that the physical chemistry of the intermediate state can be understood by the following explanation relating to proteinaceous active agents. Proteins can exist in stable intermediate conformations that are structurally and energetically distinct from either the native state or the denatured state. The inherent stability of any conformation(s) of any protein is reflected in the Gibbs free energy of the conformation(s). The Gibbs free energy for any state of a monomeric protein is described thermodynamically by the following relationship: [0075]
  • ΔG 0 =ΔH 0(T R)−TΔS 0(T R)+ΔCp 0((T−T R)−T In(T/T R))   (1)
  • where T is the temperature, T[0076] R is a reference temperature, ΔH0(TR) and TΔS0(TR) are the relative enthalpy and entropy of this state at the reference temperature, and ΔCp0 is the relative heat capacity of this state. It is convenient to chose the native state as the reference state to express all relative thermodynamic parameters.
  • The sum of the statistical weights of all states accessible to the protein is defined as the partition function Q: [0077] Q = i = 0 n - Δ Gi / RT ( 2 )
    Figure US20010039258A1-20011108-M00001
  • [0078] Equation 2 can also be written as Q = 1 + n - 1 - DGi / RT + - D Gn / RT ( 3 )
    Figure US20010039258A1-20011108-M00002
  • where the second term includes all the intermediates that become populated during the transition. The first and last terms of equation (3) are the statistical weights of the native and denatured states, respectively. Under most conditions, protein structure could be approximated by a two-state transition function: [0079]
  • Q≈1+e −ΔGn/RT   (4)
  • See, Tanford, C., [0080] Advances in Protein Chemistry, 1968, 23, 2-95. Conformations of proteins that are intermediate between the native state and the denatured state can be detected by, for example, NMR, calorimetry, and fluorescence. Dill, K. A., Shortle, D., Annu. Rev. Biochem. 60, 1991, 795-825.
  • All thermodynamic parameters can be expressed in terms of the partition function. Specifically the population of molecules in state i is given in equation (5) [0081] Pi = - Δ Gi / RT Q ( 5 )
    Figure US20010039258A1-20011108-M00003
  • Therefore, measurement of the appropriate terms in equation (1) that would allow for the calculation of the Gibbs free energy would determine the extent to which any intermediate state(s) is populated to any significant degree under defined experimental conditions. This in turn indicates the role that these intermediate state(s) play in drug delivery. The more populated the intermediate state, the more efficient the delivery. [0082]
  • Active agents suitable for use in the present invention include biologically active agents and chemically active agents, including, but not limited to, fragrances, as well as other active agents such as, for example, cosmetics. [0083]
  • Biologically active agents include, but are not limited to, pesticides, pharmacological agents, and therapeutic agents. For example, biologically active agents suitable for use in the present invention include, but are not limited to, peptides, and particularly small peptides; hormones, and particularly hormones which by themselves do not or only pass slowly through the gastro-intestinal mucosa and/or are susceptible to chemical cleavage by acids and enzymes in the gastrointestinal tract; polysaccharides, and particularly mixtures of muco-polysaccharides; carbohydrates; lipids; or any combination thereof. Further examples include, but are not limited to, human growth hormones; bovine growth hormones; growth releasing hormones; interferons; interleukin-1; insulin; heparin, and particularly low molecular weight heparin; calcitonin; erythropoietin; atrial naturetic factor; antigens; monoclonal antibodies; somatostatin; adrenocorticotropin, gonadotropin releasing hormone; oxytocin; vasopressin; cromolyn sodium (sodium or disodium chromoglycate); vancomycin; desferrioxamine (DFO); anti-microbials, including, but not limited to anti-fungal agents; or any combination thereof. [0084]
  • The methods and compositions of the present invention may combine one or more active agents. [0085]
  • Perturbants [0086]
  • Perturbants serve two purposes in the present invention. In a first embodiment, the active agent is contacted with an amount of perturbant which reversibly transforms the active agent from the native state to the intermediate state suitable for administration. The perturbant, in the appropriate amount, non-covalently complexes with the active agent to form a supramolecular complex which can be administered by a selected route. This supramolecular complex can be used as a template for the design of a mimetic or can be used as a delivery composition itself. The perturbant, in effect, fixes the active agent in the transportable intermediate state. The perturbant can be released from the supramolecular complex, such as by dilution in the circulatory system, so that the active agent can return to the native state appropriate for the route of delivery. Preferably, these perturbants have at least one hydrophilic (i.e. readily soluble in water, such as for example, a carboxylate group) and at least one hydrophobic moiety (i.e. readily soluble in an organic solvent such as, for example, a benzene group), and have a molecular weight ranging from about 150 to about 600 daltons and most preferably from about 200 to about 500 daltons. [0087]
  • Complexing perturbant compounds include, but are not limited to proteinoids including linear, non-linear, and cyclic proteinoids; modified (acylated or sulfonated) amino acids, poly amino acids, and peptides; modified amino acid, poly amino acid, or peptide derivatives (ketones or aldehydes); diketopiperazine/amino acid constructs; carboxylic acids; and various other perturbants discussed below. [0088]
  • Again without being bound by any theory, applicant believes that the non-covalent complexing may be effected by intermolecular forces including but not limited to, hydrogen bonding, hydrophilic interactions, electrostatic interactions, and Van der Waals interactions. For any given active agent/perturbant supramolecular complex, there will exist some combination of the aforementioned forces that maintain the association. [0089]
  • The association constant K[0090] a between the perturbant and the active agent can be defined according to equation (6)
  • Ka=e−ΔG/RT   (6)
  • The dissociation constant K[0091] d is the reciprocal of Ka. Thus measurement of the association constants between perturbant and active agent at a defined temperature will yield data on the molar Gibbs free energy which allows for the determination of the associated enthalpic and entropic effects. Experimentally these measurements can be made, for example, using NMR, fluorescence or calorimetry.
  • This hypothesis can be illustrated with proteins in the following manner: [0092]
  • Protein unfolding can be described according to the equilibrium that exists between its various conformational states, e.g. [0093] N k 1 I D k 2 ( 7 )
    Figure US20010039258A1-20011108-M00004
  • where N is the native state, I is the intermediate state(s), D is the denatured state, and k[0094] 1 and k2 are the respective rate constants. K1 and K2 are the respective equilibrium constants. Accordingly, Q = i = 0 n - Δ Gi / RT ( 2 )
    Figure US20010039258A1-20011108-M00005
     =1+e −ΔG 1 /R T +e −ΔG 2 /R T= 1+K 1 +K 2   (8)
  • =1+k 1 +k 1 k 2   (9)
  • This suggests that increasing the partition function of the intermediate state(s) should have a positive impact on the ability to deliver the active agent, i.e. [0095] P i = K 1 ( 1 + K 1 + K 2 ) ( 10 )
    Figure US20010039258A1-20011108-M00006
  • Because complexing must be reversible, the complexing of the perturbant with the active agent, as measured by the K[0096] a, must be strong enough to insure delivery of the drug either to the systemic circulation and/or to the target(s), but not so strong so that disengagement of the perturbant will not occur in a timely manner to allow the active agent to renature if necessary to produce the desired effect(s).
  • In a second embodiment, amounts of perturbants appropriate for the route of delivery reversibly transform the active agent to the intermediate state so that the conformation of that state can be used as a template for the preparation of mimetics. Perturbants for this purpose need not, but may, complex with the active agent. Therefore, in addition to the complexing perturbants discussed above, perturbants that change the pH of the active agent or its environment, such as for example, strong acids or strong bases; detergents; perturbants that change the ionic strength of the active agent or its environment; other agents such as for example, guanidine hydrochloride; and temperature can be used to transform the active agent. Either the supramolecular complex or the reversible intermediate state can be used as a template for mimetic design. [0097]
  • Complexing Perturbants [0098]
  • Amino acids are the basic materials used to prepare many of the complexing perturbants useful in the present invention. An amino acid is any carboxylic acid having at least one free amine group and includes naturally occurring and synthetic amino acids. The preferred amino acids for use in the present invention are α-amino acids, and most preferably are naturally occurring α-amino acids. Many amino acids and amino acid esters are readily available from a number of commercial sources such as Aldrich Chemical Co. (Milwaukee, Wis., U.S.A.); Sigma Chemical Co. (St. Louis, Mo., U.S.A.); and Fluka Chemical Corp. (Ronkonkoma, N.Y., U.S.A.). [0099]
  • Representative, but not limiting, amino acids suitable for use in the present invention are generally of the formula [0100]
    Figure US20010039258A1-20011108-C00001
  • wherein: [0101]
  • R[0102] 1 is hydrogen, C1-C4 alkyl, or C2-C4 alkenyl;
  • R[0103] 2 is C1-C24 alkyl, C2-C24 alkenyl, C3-C10 cycloalkyl, C3-C10 cycloalkenyl, phenyl, naphthyl, (C1-C10 alkyl) phenyl, (C2-C10 alkenyl) phenyl, (C1-C10 alkyl) naphthyl, (C2-C10 alkenyl) naphthyl, phenyl (C1-C10 alkyl), phenyl (C2-C10 alkenyl), naphthyl (C1-C10 alkyl), or naphthyl (C2-C10 alkenyl);
  • R[0104] 2 being optionally substituted with C1-C4 alkyl, C2-C4 alkenyl, C1-C4 alkoxy, —OH, —SH, —CO2R3, C3-C10 cycloalkyl, C3-C10 cycloalkenyl, heterocycle having 3-10 ring atoms wherein the hetero atom is one or more of N, O, S, or any combination thereof, aryl, (C1-C10 alk)aryl, ar(C1-C10 alkyl) or any combination thereof;
  • R[0105] 2 being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
  • R[0106] 3 is hydrogen, C1-C4 alkyl, or C2-C4 alkenyl.
  • The preferred naturally occurring amino acids for use in the present invention as amino acids or components of a peptide are alanine, arginine, asparagine, aspartic acid, citrulline, cysteine, cystine, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, ornithine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, hydroxy proline, γ-carboxyglutamate, phenylglycine, or O-phosphoserine. The preferred amino acids are arginine, leucine, lysine, phenylalanine, tyrosine, tryptophan, valine, and phenylglycine. [0107]
  • The preferred non-naturally occurring amino acids for use in the present invention are β-alanine, α-amino butyric acid, γ-amino butyric acid, γ-(aminophenyl) butyric acid, α-amino isobutyric acid, citrulline, ε-amino caproic acid, 7-amino heptanoic acid, β-aspartic acid, aminobenzoic acid, aminophenyl acetic acid, aminophenyl butyric acid, γ-glutamic acid, cysteine (ACM), ε-lysine, ε-lysine (A-Fmoc), methionine sulfone, norleucine, norvaline, ornithine, d-ornithine, p-nitro-phenylalanine, hydroxy proline, 1,2,3,4,-tetrahydroisoquinoline-3-carboxylic acid, and thioproline. [0108]
  • Poly amino acids are either peptides or two or more amino acids linked by a bond formed by other groups which can be linked, e.g., an ester, anhydride or an anhydride linkage. Special mention is made of non-naturally occurring poly amino acids and particularly non-naturally occurring hetero-poly amino acids, i.e. of mixed amino acids. [0109]
  • Peptides are two or more amino acids joined by a peptide bond. Peptides can vary in length from di-peptides with two amino acids to polypeptides with several hundred amino acids. See, Walker, [0110] Chambers Biological Dictionary, Cambridge, England: Chambers Cambridge, 1989, page 215. Special mention is made of non-naturally occurring peptides and particularly non-naturally occurring peptides of mixed amino acids. Special mention is also made of di-peptides tri-peptides, tetra-peptides, and penta-peptides, and particularly, the preferred peptides are di-peptides and tri-peptides. Peptides can be homo- or hetero-peptides and can include natural amino acids, synthetic amino acids, or any combination thereof.
  • Proteinoid Complexing Perturbants [0111]
  • Proteinoids are artificial polymers of amino acids. The proteinoids preferably are prepared from mixtures of amino acids. Preferred proteinoids are condensation polymers, and most preferably, are thermal condensation polymers. These polymers may be directed or random polymers. Proteinoids can be linear, branched, or cyclical, and certain proteinoids can be units of other linear, branched, or cyclical proteinoids. [0112]
  • Special mention is made of diketopiperazines. Diketopiperazines are six member ring compounds. The ring includes two nitrogen atoms and is substituted at two carbons with two oxygen atoms. Preferably, the carbonyl groups are at the 1 and 4 ring positions. These rings can be optionally, and most often are, further substituted. [0113]
  • Diketopiperazine ring systems may be generated during thermal polymerization or condensation of amino acids or amino acid derivatives. (Gyore, J; Ecet M. [0114] Proceedings Fourth ICTA (Thermal Analysis), 1974, 2, 387-394 (1974)). These six membered ring systems were presumably generated by intra-molecular cyclization of the dimer prior to further chain growth or directly from a linear peptide (Reddy, A. V., Int. J. Peptide Protein Res., 40, 472-476 (1992); Mazurov, A. A. et al., Int. J. Peptide Protein Res., 42, 14-19 (1993)).
  • Diketopiperazines can also be formed by cyclodimerization of amino acid ester derivatives as described by Katchalski et al., [0115] J. Amer. Chem. Soc., 68, 879-880 (1946), by cyclization of dipeptide ester derivatives, or by thermal dehydration of amino acid derivatives and high boiling solvents as described by Kopple et al., J. Org. Chem., 33 (2), 862-864 (1968).
  • In a typical synthesis of a diketopiperazine, the COOH group(s) of an amino acid benzyl ester are activated in a first step to yield a protected ester. The amine is deprotected and cyclized via dimerization in a second step, providing a diketopiperazine di-ester. Finally, the COOH group(s) are deprotected to provide the diketopiperazine. [0116]
  • Diketopiperazines typically are formed from α-amino acids. Preferably, the α-amino acids of which the diketopiperazines are derived are glutamic acid, aspartic acid, tyrosine, phenylalanine, and optical isomers of any of the foregoing. [0117]
  • Special mention is made of diketopiperazines of the formula [0118]
    Figure US20010039258A1-20011108-C00002
  • wherein R[0119] 4, R5, R6, and R7 independently are hydrogen, C1-C24 alkyl, C1-C24 alkenyl, phenyl, naphthyl, (C1-C10 alkyl)phenyl, (C1-C10 alkenyl)phenyl, (C1-C10 alkyl)naphthyl, (C1-C10 alkenyl)naphthyl, phenyl (C1-C10 alkyl), phenyl(C1-C10 alkenyl), naphthyl (C1-C10 alkyl), and naphthyl (C1-C10 alkenyl); any of R4, R5, R6, and R7 independently may optionally be substituted with C1-C4 alkyl, C1-C4 alkenyl, C1-C4 alkoxy, —OH, —SH, and —CO2R8 or any combination thereof; R8 is hydrogen, C1-C4 alkyl or C1-C4 alkenyl; and any of R4, R5, R6, and R7 independently may optionally be interrupted by oxygen, nitrogen, sulfur, or any combination thereof.
  • The phenyl or naphthyl groups may optionally be substituted. Suitable, but non-limiting, examples of substituents are C[0120] 1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkoxy, —OH, —SH, or CO2R9 wherein R9 is hydrogen, C1-C6 alkyl, or C1-C6 alkenyl.
  • Preferably, R[0121] 6 and R7 independently are hydrogen, C1-C4 alkyl or C1-C4 alkenyl. Special mention is made of diketopiperazines which are preferred complexing perturbants. These diketopiperazines include the unsubstituted diketopiperazine in which R4, R5, R6, and R7 are hydrogen, and diketopiperazines which are substituted at one or both of the nitrogen atoms in the ring, i.e. mono or di-N-substituted. Special mention is made of the N-substituted diketopiperazine wherein one or both of the nitrogen atoms is substituted with a methyl group.
  • Special mention is also made of diketopiperizines of the formula [0122]
    Figure US20010039258A1-20011108-C00003
  • wherein R[0123] 10 and R11 independently are hydrogen, C1-C24 alkyl, C1-C24 alkenyl, phenyl, naphthyl, (C1-C10 alkyl)phenyl, (C1-C10 alkenyl)phenyl, (C1-C10 alkyl)naphthyl, (C1-C10 alkenyl)naphthyl, phenyl (C1-C10 alkyl), phenyl(C1-C10 alkenyl), naphthyl (C1-C10 alkyl), and naphthyl (C1-C10 alkenyl); but both R10 and R11 can not be hydrogen; either or both R10 or R11 independently may optionally be substituted with C1-C4 alkyl, C1-C4 alkenyl, C1-C4 alkoxy, —OH, —SH, and —CO2R12 or any combination thereof; R12 is hydrogen, C1-C4 alkyl or C1-C4 alkenyl; and either or both R10 and R11 independently may optionally be interrupted by oxygen, nitrogen, sulfur, or any combination thereof.
  • The phenyl or naphthyl groups may optionally be substituted. Suitable, but non-limiting, examples of substituents are C[0124] 1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkoxy, —OH, —SH, or CO2R13 wherein R13 is hydrogen, C1-C6 alkyl, or C1-C6 alkenyl. When one of R10 or R11 is hydrogen, the diketopiperazine is mono-carbon-(C)-substituted. When neither R10 nor R11 is hydrogen, the diketopiperazine is di-carbon-(C)-substituted.
  • Preferably, R[0125] 10, R11, or both R10 and R11, contain at least one functional group, a functional group being a non-hydrocarbon portion responsible for characteristic reactions of the molecule. Simple functional groups are heteroatoms including, but not limited to halogens, oxygen, sulfur, nitrogen, and the like, attached to, the carbon of an alkyl group by a single or multiple bond. Other functional groups include, but are not limited to, for example, hydroxyl groups, carboxyl groups, amide groups, amine groups, substituted amine groups, and the like.
  • Preferred diketopiperazines are those which are substituted at one or two of the carbons of the ring with a functional group that includes at least one carboxyl functionality. [0126]
  • Amino Acid(s)/Diketopiperazine Complexing Perturbants
  • Diketopiperazines may also be polymerized with additional amino acids to form constructs of at least one amino acid or an ester or an amide thereof and at least one diketopiperazine, preferably covalently bonded to one another. [0127]
  • When the diketopiperazine is polymerized with additional amino acids, one or more of the R groups must contain at least one functional group, a functional group being a non-hydrocarbon portion responsible for characteristic reactions of the molecule. Simple functional groups are heteroatoms including, but not limited to halogens, oxygen, sulfur, nitrogen, and the like, attached to, the carbon of an alkyl group by a single or multiple bond. Other functional groups include, but are not limited to, for example, hydroxyl groups, carboxyl groups, amide groups, amine groups, substituted amine groups, and the like. [0128]
  • Special mention is also made of diketopiperazines which are preferred components of the amino acids/diketopiperazine perturbants of the present invention. Such preferred diketopiperazines are those which are substituted at one or two of the carbons of the ring and preferably are substituted with a functional group that includes at least one carboxyl functionality. [0129]
  • Most preferably, the diketopiperazines in the amino acids/diketopiperazine perturbants are prepared from trifunctional amino acids such as L-glutamic acid and L-aspartic acid which cyclize to form diketopiperazines. [0130]
  • The diketopiperazines can generate a bis-carboxylic acid platform which can be further condensed with other amino acids to form the perturbant. Typically, the diketopiperazine will react and covalently bond with one or more of the amino acids through the functional group(s) of the R groups of the diketopiperazines. These unique systems, because of the cis-geometry imparted by the chiral components of the diketopiperazine ring (Lannom, H. K. et al., [0131] Int. J. Peptide Protein Res., 28, 67-78 (1986)), provide an opportunity to systematically alter the structure of the terminal amino acids while holding the orientation between them fixed relative to non-cyclic analogs (Fusaoka et al., Int. J. Peptide Protein Res., 34, 104-110 (1989); Ogura, H. et al., Chem. Pharma. Bull., 23, 2474-2477 (1975). See also, Lee, B. H. et al. J. Org. Chem., 49, 2418-2423 (1984); Buyle, R., Helv. Chim. Acta, 49, 1425, 1429 (1966). Other methods of polymerization known to those skilled in the art may lend themselves to amino acid/diketopiperazine polymerization as well.
  • The amino acids/diketopiperazine perturbants may include one or more of the same or different amino acids as well as one or more of the same or different diketopiperazines as described above. [0132]
  • Ester and amide derivatives of these amino acids/diketopiperazine perturbants are also useful in the present invention. [0133]
  • Modified Amino Acid Complexing Perturbants
  • Modified amino acids, poly amino acids or peptides are either acylated or sulfonated and include amino acid amides and sulfonamides. [0134]
  • Modified amino acids are typically prepared by modifying the amino acid or an ester thereof. Many of these compounds are prepared by acylation or sulfonation with agents having the formula [0135]
  • X—Y—R4
  • wherein: [0136]
  • R[0137] 4 is the appropriate radical to yield the modification indicated in the final product,
  • Y is [0138]
    Figure US20010039258A1-20011108-C00004
  • , and X is a leaving group. Typical leaving groups include, but are not limited t, halogens such as, for example, chlorine, bromine, and iodine. Additionally, the corresponding anhydrides are modifying agents. [0139]
  • Acylated Amino Acid Complexing Perturbants
  • Special mention is made of acylated amino acids having the formula [0140]
  • Ar—Y—(R14)n—OH   IV
  • wherein [0141]
  • Ar is a substituted or unsubstituted phenyl or naphthyl; [0142]
  • Y is [0143]
    Figure US20010039258A1-20011108-C00005
  • , R[0144] 14 has the formula
    Figure US20010039258A1-20011108-C00006
  • , wherein: [0145]
  • R[0146] 15 is C1 to C24 alkyl, C1 to C24 alkenyl, phenyl, naphthyl, (C1 to C10 alkyl) phenyl, (C1 to C10 alkenyl) phenyl, (C1 to C10 alkyl) naphthyl, (C1 to C10 alkenyl) naphthyl, phenyl (C1 to C10 alkyl), phenyl (C1 to C10 alkenyl), naphthyl (C1 to C10 alkyl) and naphthyl (C1 to C10 alkenyl);
  • R[0147] 15 is optionally substituted with C1 to C4 alkyl, C1 to C4 alkenyl, C1 to C4 alkoxy, —OH, —SH and —CO2R5, cycloalkyl, cycloalkenyl, heterocyclic alkyl, alkaryl, heteroaryl, heteroalkaryl, or any combination thereof;
  • R[0148] 17 is hydrogen, C1 to C4 alkyl or C1 to C4 alkenyl;
  • R[0149] 15 is optionally interrupted by oxygen, nitrogen, sulfur or any combination thereof; and
  • R[0150] 16 is hydrogen, C1 to C4 alkyl or C1 to C4 alkenyl.
  • Special mention is also made of those having the formula [0151]
    Figure US20010039258A1-20011108-C00007
  • wherein: [0152]
  • R[0153] 18 is
  • (i) C[0154] 3-C10 cycloalkyl, optionally substituted with C1-C7 alkyl, C2-C7 alkenyl, C1-C7 alkoxy, hydroxy, phenyl, phenoxy or —CO2R21, wherein R1 is hydrogen, C1-C4 alkyl, or C2-C4 alkenyl; or
  • (ii) C[0155] 1-C6 alkyl substituted with C3-C10 cycloalkyl;
  • R[0156] 19 is hydrogen, C1-C4 alkyl, or C2-C4 alkenyl;
  • R[0157] 20 is C1-C24 alkyl, C2-C24 alkenyl, C3-C10 cycloalkyl, C3-C10 cycloalkenyl, phenyl, naphthyl, (C1-C10 alkyl) phenyl, (C2-C10 alkenyl) phenyl, (C1-C10 alkyl) naphthyl, (C2-C10 alkenyl) naphthyl, phenyl (C1-C10 alkyl), phenyl (C2-C10 alkenyl), naphthyl (C1-C10 alkyl) or naphthyl (C2-C10 alkenyl);
  • R[0158] 20 being optionally substituted with C1-C4 alkyl, C2-C4 alkenyl, C1-C4 alkoxy, —OH, —SH, —CO2R22, C3-C10 cycloalkyl, C3-C10 cycloalkenyl, heterocycle having 3-10 ring atoms wherein the hetero atom is one or more of N, O, S or any combination thereof, aryl, (C1-C10 alk)aryl, ar(C1-C10 alkyl), or any combination thereof;
  • R[0159] 20 being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
  • R[0160] 22 is hydrogen, C1-C4 alkyl, or C2-C4 alkenyl.
  • Some preferred acylated amino acids include salicyloyl phenylalanine, and the compounds having the formulas: [0161]
    Figure US20010039258A1-20011108-C00008
    A
    Figure US20010039258A1-20011108-C00009
    Compound n m X
    XXXVIIA
    0 0 4-Cl
    XXXVIIIA
    3 0 H
    XXIXA
    3 1 4-CH3
    XLA 3 1 2-F
    XLIA
    3 1 2-CH3
    XLIIA 3 0 3-CF3
    XLIIIA 3 4 H
    XLIVA
    3 0 3-Cl
    XLVA
    3 0 3-F
    XLVIA
    3 0 3-CH3
    XLVIIA 0 0 2-CF3
    XLVIIIA 1 2 H
    XLIXA
    3 2 2-F
    LA
    3 0 3,4-OCH2O—
    LIA 3 0 2-COOH
    LIIA
    1 0 2-OH
    LIIIA
    3 0 2,6-dihydroxy
    LIVA
    2 0 2-OH
    LVA
    0 0 2,4-difluoro
    LVIA
    2 0 2,6-dihydroxy
    LVIIA
    0 0 4-CF3
    LVIIIA 3 0 3-NMe2
    LIXA 2 0 3-NMe2
    LXA 3 0 2,6-dimethyl
    LXIA
    3 0 2-NO2
    LXIIA 3 0 2-CF3
    LXIIIA 3 0 4-n-Pr
    LXIVA
    3 0 2-NH2
    LXVA 3 0 2-OCH3
    LXVIA 3 0 3-NO2
    LXVIIA 3 0 3-NH2
    LXVIIIA 2 0 2-NO2
    LXIXA 2 0 2-NH2
    LXXA 3 0 2-OCF3
    LXXIA 2 0 2-OCH3
    LXXIIA 2 0 2-OCF3
  • [0162]
    B
    Figure US20010039258A1-20011108-C00010
    Compound n X
    LXXIIIB
    3 4-CF3
    LXXIVB 1 2-F
    LXXVB
    1 4-CF3
    LXXVIB 3 3,4-dimethoxy
    LXXVIIB
    0 3-OCH3
    LXXVIIIB 3 3-OCH3
    LXXIXB 3 2,6-difluoro
    LXXXB
    3 4-CH3
    LXXXIB 1 4-OCH3
    LXXXIIB 2 2-F
    LXXXIIIB
    0 2-F
    LXXXIVB
    2 4-OCH3
    LXXXVB 0 2-OCH3
    LXXXVIB 2 2-OCH3
    LXXXVIIB 0 4-CF3
    LXXXVIIIB 3 3-F
    LXXXIXB
    3 2-OCH3
  • [0163]
    C
    Figure US20010039258A1-20011108-C00011
    Compound n m X
    VII-C 3 0 2-carboxycyclohexyl
    VIII-C 3 3 cyclohexyl
    IX-C 3 0 2-adamantyl
    X-C 3 0 1-morpholino
  • [0164]
    D
    Figure US20010039258A1-20011108-C00012
    Compound m
    XI-D 0
    XII-D 3
  • [0165]
    E
    Figure US20010039258A1-20011108-C00013
    Compound X
    XIII-E OH
    XIV-E ═O
  • [0166]
    F
    Figure US20010039258A1-20011108-C00014
    Compound n
    XV-F 0
    XVI-F 2
  • [0167]
    Figure US20010039258A1-20011108-C00015
    G
    Figure US20010039258A1-20011108-C00016
    Compound n m X
    CXI-G 6 0 2-OH
    CXII-G 7 3 H
    CXIII-G 7 0 2-I
    CXIV-G 7 0 2-Br
    CXV-G 7 0 3-NO2
    CXVI-G 7 0 3-N(CH3)2
    CXVII-G 7 0 2-NO2
    CXVIII-G 7 0 4-NO2
    CXIX-G 9 0 2-OH
  • [0168]
    H
    Figure US20010039258A1-20011108-C00017
    Compound X
    CXX-H 1-morpholino
    CXXI-H O-t-Butyl
    CXXII-H CH(CH2Ph)NC(O)O-t-Bu
    CXXIII-H 2-hydroxyphenyl
  • organic acid compounds, and their salts, having an aromatic amide group, having a hydroxy group substituted in the ortho position on the aromatic ring, and a lipophilic chain with from about 4 carbon atoms to about 20 atoms in the chain are also useful as perturbants. In a preferred form the lipophilic chain can have from 5 to 20 carbon atoms. [0169]
  • Perturbants which are also useful also include those having the formula [0170]
  • 2—HO—Ar—CONR8—R7—COOH
  • wherein [0171]
  • Ar is a substituted or unsubstituted phenyl or naphthyl; [0172]
  • R[0173] 7 is selected from the group consisting of C4 to C20 alkyl, C4 to C20 alkenyl, phenyl, naphthyl, (C1 to C10 alkyl) phenyl, (C1 to C10 alkenyl) phenyl, (C1 to C10 alkyl) naphthyl, (C1 to C10 alkenyl) naphthyl, phenyl (C1 to C10 alkyl), phenyl (C1 to C10 alkenyl), naphthyl (C1 to C10 alkyl), and naphthyl (C1 to C10 alkenyl);
  • R[0174] 8 is selected from the group consisting of hydrogen, C1 to C4 alkyl, C1 to C4 alkenyl, hydroxy, and C1 to C4 alkoxy;
  • R[0175] 7 is optionally substituted with C1 to C4 alkyl, C1 to C4 alkenyl, C1 to C4 alkoxy, —OH, —SH and —CO2R9 or any combination thereof;
  • R[0176] 9 is hydrogen, C1 to C4 alkyl or C1 to C4 alkenyl;
  • R[0177] 7 is optionally interrupted by oxygen, nitrogen, sulfur or any combination thereof;
  • with the proviso that the compounds are not substituted with an amino group in the position alpha to the acid group, or salts thereof. [0178]
  • The preferred R[0179] 6 groups are of C4 to C20 alkyl and C4 to C20 alkenyl. The most preferred R6 groups are C5 to C20 alkyl and C5 to C20 alkenyl.
  • A preferred carrier compound can have the formula [0180]
    Figure US20010039258A1-20011108-C00018
  • wherein R[0181] 7 is defined above.
  • Special mention is made of compounds having the formula: [0182]
    Figure US20010039258A1-20011108-C00019
  • wherein A is Try, Leu, Arg, Trp, or Cit; and [0183]
  • optionally wherein if A is Try, Arg, Trp or Cit; A is acylated at 2 or more functional groups. [0184]
  • Preferred compounds also include those wherein A is Try; A is Tyr and is acylated at 2 functional groups; A is Leu; A is Arg; A is Arg and is acylated at 2 functional groups; A is Trp; A is Trp and is acylated at 2 functional groups; A is Cit; and A is Cit and is acylated at 2 functional groups. [0185]
  • Special mention is also made of compounds having the formula: [0186]
    Figure US20010039258A1-20011108-C00020
  • wherein A is Arg or Leu; and [0187]
  • wherein if A is Arg, A is optionally acylated at 2 or more functional groups; [0188]
    Figure US20010039258A1-20011108-C00021
  • where A is Leu or phenylglycine; [0189]
    Figure US20010039258A1-20011108-C00022
  • wherein A is phenylglycine; and [0190]
    Figure US20010039258A1-20011108-C00023
  • wherein A is phenylglycine. [0191]
  • Acylated amino acids may be prepared by reacting single amino acids, mixtures of two or more amino acids, or amino acid esters with an amine modifying agent which reacts with free amino moieties present in the amino acids to form amides. [0192]
  • Suitable, but non-limiting, examples of acylating agents useful in preparing acylated amino acids include acid chloride acylating agents having the formula [0193]
    Figure US20010039258A1-20011108-C00024
  • wherein: [0194]
  • R[0195] 23 an appropriate group for the modified amino acid being prepared, such as, but not limited to, alkyl, alkenyl, cycloalkyl, or aromatic, and particularly methyl, ethyl, cyclohexyl, cyclophenyl, phenyl, or benzyl, and X is a leaving group. Typical leaving groups include, but are not limited to, halogens such as chlorine, bromine and iodine.
  • Examples of the acylating agents include, but are not limited to, acyl halides including, but not limited to, acetyl chloride, propyl chloride, cyclohexanoyl chloride, cyclopentanoyl chloride, and cycloheptanoyl chloride, benzoyl chloride, hippuryl chloride and the like; and anhydrides, such as acetic anhydride, propyl anhydride, cyclohexanoic anhydride, benzoic anhydride, hippuric anhydride and the like. Preferred acylating agents include benzoyl chloride, hippuryl chloride, acetyl chloride, cyclohexanoyl chloride, cyclopentanoyl chloride, and cycloheptanoyl chloride. [0196]
  • The amine groups can also be modified by the reaction of a carboxylic acid with coupling agents such as the carbodiimide derivatives of amino acids, particularly hydrophilic amino acids such as phenylalanine, tryptophan, and tyrosine. Further examples include dicyclohexylcarbodiimide and the like. [0197]
  • If the amino acid is multifunctional, i.e. has more than one —OH, —NH[0198] 2 or —SH group, then it may optionally be acylated at one or more functional groups to form, for example, an ester, amide, or thioester linkage.
  • For example, in the preparation of many acylated amino acids, the amino acids are dissolved in an aqueous alkaline solution of a metal hydroxide, e.g., sodium or potassium hydroxide and the acylating agent added. The reaction time can range from about 1 hour and about 4 hours, preferably about 2-2.5 hours. The temperature of the mixture is maintained at a temperature generally ranging between about 5° C. and about 70° C., preferably between about 10° C. and about 50° C. The amount of alkali employed per equivalent of NH[0199] 2 groups in the amino acids generally ranges between about 1.25 moles and about 3 moles, and is preferably between about 1.5 moles and about 2.25 moles per equivalent of NH2. The pH of the reaction solution generally ranges between about pH 8 and about pH 13, and is preferably between about pH 10 and about pH 12. The amount of amino modifying agent employed in relation to the quantity of amino acids is based on the moles of total free NH2 in the amino acids. In general, the amino modifying agent is employed in an amount ranging between about 0.5 and about 2.5 mole equivalents, preferably between about 0.75 and about 1.25 equivalents, per molar equivalent of total NH2 groups in the amino acids.
  • The modified amino acid formation reaction is quenched by adjusting the pH of the mixture with a suitable acid, e.g., concentrated hydrochloric acid, until the pH reaches between about 2 and about 3. The mixture separates on standing at room temperature to form a transparent upper layer and a white or off-white precipitate. The upper layer is discarded, and modified amino acids are collected by filtration or decantation. The crude modified amino acids are then mixed with water. Insoluble materials are removed by filtration and the filtrate is dried in vacuo. The yield of modified amino acids generally ranges between about 30 and about 60%, and usually about 45%. The present invention also contemplates amino acids which have been modified by multiple acylation, e.g., diacylation or triacylation. [0200]
  • If desired, amino acid esters, such as, for example benzyl, methyl, or ethyl esters of amino acid compounds, may be used to prepare the modified amino acids of the invention. The amino acid ester, dissolved in a suitable organic solvent such as dimethylformamide, pyridine, or tetrahydrofuran is reacted with the appropriate amino modifying agent at a temperature ranging between 5° C. and about 70° C., preferably about 25° C., for a period ranging between about 7 and about 24 hours. The amount of amino acid modifying agent used relative to the amino acid ester is the same as described above for amino acids. This reaction may be carried out with or without a base such as, for example, triethylamine or diisopropylethylamine. [0201]
  • If amino acid esters or amides are the starting materials, they are dissolved in a suitable organic solvent such as dimethylformamide or pyridine, are reacted with the amino modifying agent at a temperature ranging between about 5° C. and about 70° C., preferably about 25° C., for a period ranging between about 7 and about 24 hours. The amount of amino modifying agents used relative to the amino acid esters are the same as described above for amino acids. [0202]
  • Thereafter, the reaction solvent is removed under negative pressure and optionally the ester or amide functionality can be removed by hydrolyzing the modified amino acid ester with a suitable alkaline solution, e.g., 1N sodium hydroxide, at a temperature ranging between about 50° C. and about 80° C., preferably about 70° C., for a period of time sufficient to hydrolyze off the ester group and form the modified amino acid having a free carboxyl group. The hydrolysis mixture is then cooled to room temperature and acidified, e.g., with an aqueous 25% hydrochloric acid solution, to a pH ranging between about 2 and about 2.5. The modified amino acid precipitates out of solution and is recovered by conventional means such as filtration or decantation. [0203]
  • The modified amino acids may be purified by acid precipitation, recrystallization or by fractionation on solid column supports. Fractionation may be performed on a suitable solid column supports such as silica gel, alumina, using solvent mixtures such as acetic acid/butanol/water as the mobile phase; reverse phase column supports using trifluoroacetic acid/acetonitrile mixtures as the mobile phase; and ion exchange chromatography using water as the mobile phase. The modified amino acids may also be purified by extraction with a lower alcohol such as methanol, butanol, or isopropanol to remove impurities such as inorganic salts. [0204]
  • The modified amino acids generally are soluble in alkaline aqueous solution (pH≧9.0); partially soluble in ethanol, n-butanol and 1:1 (v/v) toluene/ethanol solution and insoluble in neutral water. The alkali metal salts, e.g., the sodium salt of the derivatized amino acids are generally soluble in water at about a pH of 6-8. [0205]
  • In poly amino acids or peptides, one or more of the amino acids may be modified (acylated). Modified poly amino acids and peptides may include one or more acylated amino acid(s). Although linear modified poly amino acids and peptides will generally include only one acylated amino acid, other poly amino acid and peptide configurations can include more than one acylated amino acid. Poly amino acids and peptides can be polymerized with the acylated amino acid(s) or can be acylated after polymerization. [0206]
  • Special mention is made of the compound: [0207]
    Figure US20010039258A1-20011108-C00025
  • wherein A and B independently are Arg or Leu. [0208]
  • Sulfonated Amino Acid Complexing Perturbants
  • Sulfonated modified amino acids, poly amino acids, and peptides are modified by sulfonating at least one free amine group with a sulfonating agent which reacts with at least one of the free amine groups present. [0209]
  • Special mention is made of compounds of the formula [0210]
  • Ar—Y—(R24)n—OH   LXI
  • wherein [0211]
  • Ar is a substituted or unsubstituted phenyl or naphthyl; [0212]
  • Y is —SO[0213] 2—, R24 has the formula
    Figure US20010039258A1-20011108-C00026
  • , wherein: [0214]
  • R[0215] 25 is C1 to C24 alkyl, C1 to C24 alkenyl, phenyl, naphthyl, (C1 to C10 alkyl) phenyl, (C1 to C10 alkenyl) phenyl, (C1 to C10 alkyl) naphthyl, (C1 to C10 alkenyl) naphthyl, phenyl (C1 to C10 alkyl), phenyl (C1 to C10 alkenyl), naphthyl (C1 to C10 alkyl) and naphthyl (C1 to C10 alkenyl);
  • R[0216] 25 is optionally substituted with C1 to C4 alkyl, C1 to C4 alkenyl, C1 to C4 alkoxy, —OH, —SH and —CO2R27 or any combination thereof;
  • R[0217] 27 is hydrogen, C1 to C4 alkyl or C1 to C4 alkenyl;
  • R[0218] 25 is optionally interrupted by oxygen, nitrogen, sulfur or any combination thereof; and
  • R[0219] 26 is hydrogen, C1 to C4 alkyl or C1 to C4 alkenyl.
  • Suitable, but non-limiting, examples of sulfonating agents useful in preparing sulfonated amino acids include sulfonating agents having the formula R[0220] 28—SO2—X wherein R28 is an appropriate group for the modified amino acid being prepared such as, but not limited to, alkyl, alkenyl, cycloalkyl, or aromatics and X is a leaving group as described above. One example of a sulfonating agent is benzene sulfonyl chloride.
  • Modified poly amino acids and peptides may include one or more sulfonated amino acid(s). Although linear modified poly amino acids and peptides used generally include only one sulfonated amino acid, other poly amino acid and peptide configurations can include more than one sulfonated amino acid. Poly amino acids and peptides can be polymerized with the sulfonated amino acid(s) or can be sulfonated after polymerization. [0221]
  • Special mention is made of the compound [0222]
    Figure US20010039258A1-20011108-C00027
  • Modified Amino Acid Derivative Complexing Perturbants
  • Modified amino acid, polyamino acid, or peptide derivatives are amino acids, poly amino acids, or peptides which have had at least one acyl-terminus converted to an aldehyde or a ketone, and are acylated at at least one free amine group, with an acylating agent which reacts with at least one of the free amine groups present. [0223]
  • Amino acid, poly amino acid, or peptide derivatives can be readily prepared by reduction of amino acid esters or peptide esters with an appropriate reducing agent. For example, amino acid, poly amino acid, or peptide aldehydes can be prepared as described in an article by R. Chen et al., [0224] Biochemistry, 1979, 18, 921-926. Amino acid, poly amino acid, or peptide ketones can be prepared by the procedure described in Organic Syntheses, Col. Vol. IV, Wiley, (1963), page 5. Acylation is discussed above.
  • For example, the derivatives may be prepared by reacting a single amino acid, poly amino acid, or peptide derivative or mixtures of two or more amino acid or peptide derivatives, with an acylating agent or an amine modifying agent which reacts with free amino moieties present in the derivatives to form amides. The amino acid, poly amino acid, or peptide can be modified and subsequently derivatized, derivatized and subsequently modified, or simultaneously modified and derivatized. Protecting groups may be used to avoid unwanted side reactions as would be known to those skilled in the art. [0225]
  • In modified poly amino acid or peptide derivative, one or more of the amino acid may be derivatized (an aldehyde or a ketone) and/or modified, (acylated) but there must be at least one derivative and at least one modification. [0226]
  • Special mention is made of the modified amino acid derivatives N-cyclohexanoyl-Phe aldehyde, N-acetyl-Phe-aldehyde, N-acetyl-Tyr ketone, N-acetyl-Lys ketone and N-acetyl-Leu ketone, and N-cyclohexanoyl phenyl-alanine aldehyde. [0227]
  • Carboxylic Acid Complexing Perturbants
  • Various carboxylic acids and salts of these carboxylic acids may be used as complexing perturbants. These carboxylic acids have the formula: [0228]
  • R29—CO2H   LXII
  • wherein [0229]
  • R[0230] 29 is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2 to C10 alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
  • R[0231] 29 being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R30, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alk)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
  • R[0232] 29 being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
  • R[0233] 30 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl.
  • The preferred carboxylic acids are cyclohexanecarboxylic acid, cyclopentanecarboxylic acid, cycloheptanecarboxylic acid, hexanoic acid, 3-cyclohexanepropanoic acid, methylcyclohexanecarboxylic acid, 1,2-cyclohexanedicarboxylic acid, 1,3-cyclohexanedicarboxylic acid, 1,4-cyclohexanedicarboxylic acid, 1-adamantanecarboxylic acid, phenylpropanoic acid, adipic acid, cyclohexanepentanoic acid, cyclohexanebutanoic acid, pentylcyclohexanoic acid, 2-cyclopentanehexanoic acid, cyclohexane pentanoic acid, hexanedioic acid, cyclohexanebutanoic acid, and (4-methylphenyl) cyclohexane acetic acid. [0234]
  • Other Examples of Complexing Perturbants
  • Although all complexing perturbants which can form the supramolecular complexes described herein are within the scope of the present invention, other examples of complexing perturbants include, but are not limited to, 2-carboxymethyl-phenylalanine-leucine; 2-benzyl succinic acid, an actinonin, phenylsulfonyl aminophenyl-butyric acid, [0235]
    Figure US20010039258A1-20011108-C00028
  • Mimetics [0236]
  • Mimetics within the scope of the present invention are constructs which are structural and/or functional equivalents of an original entity. Structural and chemically functional mimetics of the supramolecular complexes and the reversible transportable intermediate states of active agents are not necessarily peptidic, as non-peptidic mimetics can be prepared which have the appropriate chemical and/or structural properties. However, preferred mimetics are peptides which have a different primary structure than the supramolecular complex or the intermediate state, but retain the same secondary and tertiary structure of the supramolecular complex or the intermediate state. Although mimetics may have less bioactivity than a native state or intermediate state active agent or supra molecular complex, the mimetics may possess other important properties which may not be possessed by the native state such as, for example, further increased ability to be delivered orally. [0237]
  • Methods of preparation of such mimetics are described, for example, in Yamazaki et al., [0238] Chirality 3:268-276 (1991); Wiley et al., Peptidomimetics Derived From Natural Products, Medicinal Research Reviews, Vol. 13, No. 3, 327-384 (1993); Gurrath et al., Eur. J. Biochem 210:991-921 (1992); Yamazaki et al, Int. J. Peptide Protein Res. 37:364-381 (1991); Bach et al., Int. J. Peptide Protein Res. 38:314-323 (1991); Clark et al., J. Med. Chem. 32:2026-2038 (1989); Portoghese, J. Med. Chem. 34:(6) 1715-1720 (1991); Zhou et al., J. Immunol. 149 (5) 1763-1769 (Sep. 1, 1992); Holzman et al., J. Protein Chem. 10: (5) 553-563 (1991); Masler et al., Arch. Insect Biochem. and Physiol. 22:87-111 (1993); Saragovi et al., Biotechnology 10: (July 1992); Olmsteel et al., J. Med. Chem. 36:(1) 179-180 (1993); Malin et al. Peptides 14:47-51 (1993); and Kouns et al., Blood 80:(10) 2539-2537 (1992); Tanaka et al., Biophys. Chem. 50 (1994) 47-61; DeGrado et al., Science 243 (Feb. 3, 1989); Regan et al., Science 241: 976-978 (Aug. 19, 1988); Matouschek et al, Nature 340: 122-126 (Jul. 13, 1989); Parker et al., Peptide Research 4: (6) 347-354 (1991); Parker et al., Peptide Research 4:(6) 355-363 (1991); Federov et al., J. Mol. Biol. 225: 927-931 (1992); Ptitsyn et al., Biopolymers 22: 15-25 (1983); Ptitsyn et al., Protein Engineering 2:(6) 443-447 (1989).
  • For example, protein structures are determined by the collective intra- and inter-molecular interactions of the constituent amino acids. In alpha helices, the first and fourth amino acid in the helix interact non-covalently with one another. This pattern repeats through the entire helix except for the first four and last four amino acids. In addition, the side chains of amino acids can interact with one another. For example, the phenyl side chain of phenylaline would probably not be solvent exposed if that phenylalanine were found in a helix. If the interactions of that phenylalanine contributed to helix stability then substituting an alanine for a phenylalanine would disrupt the helix and change the conformation of a protein. [0239]
  • Therefore, a mimetic could be created by first determining which amino acid side chains became solvent exposed and thus removed from contributing to stabilization of the native state such as by the technique of scanning mutagenesis. Mutants containing amino acid substitutions at those same sights could be created so that the substituted amino acids would render the protein conformation more intermediate-like that native-like. Confirmation that the appropriate structure had been synthesized could come from spectroscopy and other analytical methods. [0240]
  • Delivery Compositions [0241]
  • Delivery compositions which include the supramolecular complex described above are typically formulated by mixing an effective amount of perturbant appropriate for the chosen route of delivery i.e, subcutaneous, nasal, or sublingual, with the active agent. The components can be prepared well prior to administration or can be mixed just prior to administration. [0242]
  • The delivery compositions of the present invention may also include one or more enzyme inhibitors. Such enzyme inhibitors include, but are not limited to, compounds such as actinonin or epiactinonin and derivatives thereof. These compounds have the formulas below: [0243]
    Figure US20010039258A1-20011108-C00029
  • Derivatives of these compounds are disclosed in U.S. Pat. No. 5,206,384. Actinonin derivatives have the formula: [0244]
    Figure US20010039258A1-20011108-C00030
  • wherein R[0245] 31 is sulfoxymethyl or carboxyl or a substituted carboxy group selected from carboxamide, hydroxyaminocarbonyl and alkoxycarbonyl groups; and R32 is hydroxyl, alkoxy, hydroxyamino or sulfoxyamino group. Other enzyme inhibitors include, but are not limited to, aprotinin (Trasylol) and Bowman-Birk inhibitor.
  • The stabilizing additives may be incorporated into a carrier solution of the supramolecular complex. With some drugs, the presence of such additives promotes the stability and dispersibility of the agent in solution. [0246]
  • The stabilizing 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, methyl cellulose, polyethylene glycol, carboxylic acids and salts thereof, and polylysine. The preferred stabilizing additives are gum acacia, gelatin and methyl cellulose. [0247]
  • The amount of active agent is an amount effective to accomplish the purpose of the particular active agent. The amount in the composition typically is a pharmacologically or biologically effective amount. However, the amount can be less than a pharmacologically or biologically effective amount when the composition is used in a dosage unit form, such as a capsule, a tablet or a liquid, because the dosage unit form may contain a multiplicity of carrier/biologically or chemically active agent compositions or may contain a divided pharmacologically or biologically effective amount. The total effective amounts can then be administered in cumulative units containing, in total, pharmacologically or biologically or chemically active amounts of biologically or pharmacologically active agent. [0248]
  • The total amount of active agent, and particularly biologically or chemically active agent, to be used can be determined by those skilled in the art. However, it has surprisingly been found that with some biologically or chemically active agents, the use of the presently disclosed carriers provides extremely efficient delivery, particularly in, intranasal, sublingual, or subcutaneous systems. Therefore, lower amounts of biologically or chemically active agent than those used in prior dosage unit forms or delivery systems can be administered to the subject, while still achieving the same blood levels and therapeutic effects. [0249]
  • The amount of perturbant used is a delivery effective amount. [0250]
  • The compositions of the present invention may be formulated into dosage units by the addition of one or more excipient(s), diluent(s), disintegrant(s), lubricant(s), plasticizer(s), colorant(s), or dosing vehicle(s). Preferred dosage unit forms are oral dosage unit forms. Most preferred dosage unit forms include, but are not limited to, tablets, capsules, or liquids. The dosage unit forms can include biologically, pharmacologically, or therapeutically effective amounts of the active agent or can include less than such an amount if multiple dosage unit forms are to be used to administer a total dosage of the active agent. Dosage unit forms are prepared by methods conventional in the art. [0251]
  • The subject invention is useful for administering biologically active agents to any animals such as birds; mammals, such as primates and particularly humans; and insects. The systems are particularly advantageous for delivering chemical or biologically active agents which would otherwise be destroyed or rendered less effective by conditions encountered before the active agent in the native state reaches its target zone (i.e. the area to which the active agent to be delivered) and by conditions within the body of the animal to which they are administered. Particularly, the present invention is useful in subcutaneously, intranasally, or sublingually administering active agents. [0252]
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The following examples illustrate the invention without limitation. All parts and percentages are by weight unless otherwise indicated. [0253]
  • EXAMPLE 1 α-Interferon Native Gels
  • Native gradient gels (Pharmacia) were run with 647 μg/ml of α-interferon, (Intron-A—Schering-Plough) and increasing amounts (10-500 mg/mL) of perturbant (mixture of L-Valine, L-Leucine, L-phenylalanine, L-lysine and L-arginine modified with benzenesulfonylchloride) (valine—7.4%, leucine—16.5%, phenylalanine—40.3%, lysine—16.2% and arginine—19.6%). 4 μl of material were loaded onto the gel using a 6/4 comb for loading. [0254]
  • Results are illustrated in FIG. 1. [0255]
  • [0256] Lane 1=High molecular weight marker (Bio-Rad)—1:20 dilution w/dH2O—(5 μl—>100 μl).
  • [0257] Lane 2=α-interferon A (647 μg/mL) control 5 μl+5 μl Bromophenol Blue (BPB)—(1.29 μg loaded).
  • [0258] Lane 3=α-interferon+perturbant (10 mg/mL)−50 μl α-interferon+50 μl BPB=100 μl (1.29 μg loaded).
  • [0259] Lane 4=α-interferon+perturbant (50 mg/mL) 50 μl α-interferon+50 μl BPB=100 μl (1.29 μg loaded).
  • [0260] Lane 5=α-interferon+perturbant (100 mg/mL) 50 μl α-interferon+50 μl BPB=100 μl (1.29 μg loaded).
  • [0261] Lane 6=α-interferon+perturbant (500 mg/mL) 5 μl α-interferon+5 μl BPB=10 μl (1.29 μg loaded).
  • EXAMPLE 1A α-Interferon Native Gradient Gel
  • The method of Example 1 was followed substituting the thermal condensation product of glutamic acid, aspartic acid, tyrosine, and phenyl-alanine (Glu-Asp-Tye-Phe) that was fractionated through a 3000 molecular weight cut-off filter for the perturbant. [0262]
  • Results are illustrated in FIG. 2. [0263]
  • Samples [0264]
  • [0265] Lane 1=High Molecular Weight marker (Bio-Rad).
  • [0266] Lane 2=α-interferon (647 μg/mL)−5 μl+5 μl BPB control.
  • [0267] Lane 3=α-interferon+perturbant (10 mg/mL)−50 μl+50 μl BPB=100 μl.
  • [0268] Lane 4=α-interferon+perturbant−50 μl+50 μl BPB=100 μl.
  • [0269] Lane 5=α-interferon+perturbant (100 mg/mL)−50 μl Intron A+50 μl BPB=100 μl.
  • [0270] Lane 6=α-interferon+perturbant (500 mg/mL)−5 μl Intron A+50 μl BPB=100 μl.
  • Examples 1 and 1A illustrate that α-interferon alone ([0271] lane 2 in FIGS. 1 and 2) banded at the appropriate molecular weight (approximately 19,000 Daltons). As the amount of perturbant added is increased in each subsequent lane relative to a fixed concentration of α-interferon, the α-interferon migrates to a lower, rather than a higher, molecular weight. The change seen with the perturbant of Example 1 is more pronounced than that seen with the perturbant of Example 1A. This indicates that the α-interferon structure is changing due to the two different perturbants, because if structure were not changing, there would be a shift towards higher molecular weight as perturbant complexes with the active agent.
  • EXAMPLE 2 Isothermal Titration Calometry
  • A dosing composition of the perturbant of Example 1 at 2.4 mM and sCt at 0.3 mM was prepared, and isothermal titration calorimetry was performed at pH 6.5 and 4.5. The buffer at pH 6.5 was 30 mM Hepes-30 mM NaCl, and the buffer at pH 4.5, was 30 mM sodium acetate-30 mM NaCl. [0272]
  • All experiments were performed at 30° C. using 8.0 mM perturbant in the dropping syringe and 1.0 mM calcitonin in the calorimeter cell. In all experiments, 15×10 μl increments of perturbant were added in 10 second duration additions with 2 minutes equilibration between additions. [0273]
  • Results were validated in experiments where perturbant (8 mM) was placed in the dropping syringe, and equivalent increments were added to pH 4.5 buffer (no sCt) and where perturbant was placed in the dropping syringe and 10 μl increments were added to pH 6.5 buffer (no sCt). Titration curves were not obtained in these experiments, and the results showed that heat of mixing and/or dilution of perturbant is negligible. Therefore, the experimental isotherms were not corrected by background subtraction. [0274]
  • Results are illustrated in Table 1 below. [0275]
  • EXAMPLE 2A
  • The method of Example 2 was followed substituting the perturbant of Example 1A. Results were validated in experiments where perturbant was placed in the dropping syringe, and equivalent increments were added to pH 4.5 buffer (no sct). [0276]
  • Results are illustrated in Table 1 below. [0277]
    TABLE 1
    Binding Parameters of Perturbants
    as Determined by ITC1
    KD ΔH ΔS
    (M) (cal/mol) (cal/mol ° K) N
    pH 6.6
    Example 4 4.59 × 10−8 +240 +34.4    0.6
    Example 4A 6.99 × 10−9 +277 +38.3   11.6
    pH 4.5
    Example 4 Precipitates
    Example 4A 1.29 × 10−4 +553 +19.8  +0.8
  • [0278]
  • EXAMPLE 3 GuHCl Denaturation of α-interferon
  • A stock solution of 9.1 mg/mL of αinterferon (Schering Plough Corp.) in 20 mM sodium phosphate buffer at pH 7.2 was prepared. Samples were prepared by diluting the α-interferon with the sodium phosphate buffer and 10 M guanidine hydrochloride (GuHCl) (Sigma Chemical Co.—St. Louis, Mo.) stock solution to 200 μg/mL concentration of α-interferon at various concentrations of GuHCl. Diluted samples were allowed to come to equilibrium by incubation for approximately 30 minutes at room temperature prior to measurement. [0279]
  • Fluorescence measurements were made at 25° C. using a Hitachi F-4500. Protein tryptophan fluorescence was observed at an excitation wavelength of 298 nm and an emission wavelength of 343 nm. ANS (1-anilinonapthalene-8-sulfonate) fluorescence was observed at an excitation wavelength of 355 nm and an emission wavelength of 530 nm. For all fluorescence measurements, a 5 nm spectral bandpass was chosen for both excitation and emission. [0280]
  • Results are illustrated in FIG. 3. [0281]
  • EXAMPLE 4 Concentration Effect of GuHCl on α-Interferon Configuration
  • GuHCl 5M stock solution was prepared using 20 mM sodium phosphate, pH 7.2 buffer. After dilution, the pH of the stock was checked and adjusted by concentrated HCl. To determine the concentration of final solution the refractive index referenced in [0282] Methods in Enzymology, Vol. 6, page 43 by Yasuhiko Nozaki was used.
  • α-interferon stock (9.1 mg/mL) was mixed with sufficient amounts of GuHCl to yield the concentrations of Table 1A below: [0283]
    TABLE 1A
    α-Interferon/GuHCI Solutions
    GuHCI (M) α-IFN (mg/mL)
    0.5 0.60
    1.0 0.53
    1.5 0.60
    2.0 0.50
    3.0 0.60
    4.0 0.50
  • Differential scanning calorimetry (DSC) was run, and results are illustrated in FIG. 4. [0284]
  • EXAMPLE 5 pH Titration of Intron A as Measured by Intrinsic Tryptophan Fluorescence
  • A stock solution of 9.1 mg/mL α-interferon in 20 mM sodium phosphate buffer at pH 7.2 (Schering Plough Corp.) was prepared. Samples were prepared by diluting the α-interferon to a concentration of 200 ug/mL into solution buffered at various pH values using the following buffers: Glycine at [0285] pH 2 and 12, sodium phosphate at pH 3, 4, 5, 7, and boric acid at pH 8. These buffers were prepared as described in the Practical Handbook of Biochemistry and Molecular Biology, Edited by Gerald D. Fasman, 1990. Diluted samples were allowed to come to equilibrium by incubation for approximately 30 minutes at room temperature prior to measurement.
  • Fluorescence was measured according to the procedure of Example 3. Results are illustrated in FIG. 5. [0286]
  • EXAMPLE 6 pH Titration of Insulin Measured by ANS Fluorescence
  • A stock solution was prepared by dissolving 2 mg of insulin in 1 mL of deionized water. 1-anilinonaphthalene-8-sulphonate (ANS) stock solution was prepared by dissolving 10 mg in 10 mL of deionized water. Samples were prepared by diluting the insulin to a concentration of 200 ug/mL into solution buffered at various pH values using the following buffers: Glycine at [0287] pH 2 and 12, sodium phosphate at pH 3, 4, 5, 7, and boric acid at pH 8. These buffers were prepared as described in the Practical Handbook of Biochemistry and Molecular Biology, Edited by Gerald D. Fasman, 1990. The final ANS concentration was 90 ug/mL. Diluted samples were allowed to come to equilibrium by incubation for approx. 30 minutes at room temperature prior to measurement.
  • Fluorescence was measured according to the procedure of Example 3. Results are illustrated in FIG. 6. [0288]
  • EXAMPLE 7 Reversibility of Circular Dichroism Spectra of α-interferon at pH 2 and 7.2
  • Circular dichroism spectra of α-interferon were generated at pH 7.2. The pH of the solution was then readjusted to [0289] pH 2, and the sample was rescanned. The sample solution was then readjusted to 7.2 and rescanned.
  • Concentration of α-interferon was 9.2 μM or 0.17848 mg/mL, ([IFN] stock=9.1 mg/mL). Buffers used were 20 mM NaPhosphate at pH 7.2; and 20 mM Glycine at pH 2.0. [0290]
  • Reversal of the pH to 7.2 resulted in complete restoration of the native structure, demonstrating the reversibility of the intermediate state. It is believed that the free energy difference between the native state and the intermediate state is small. [0291]
  • Results are illustrated in FIGS. 7A and 7B. [0292]
  • EXAMPLE 8 Circular Dichroism Spectra of α-interferon at 7.2—pH Dependence
  • The extent of ordered secondary structure of α-interferon at different pH's was determined by circular dichroism (CD) measurements in the far UV range. The large dilution factor of interferon stock (˜50 times) resulted in the sample being at the proper pH. Concentration of α-interferon was 9.2 μM or 0.17848 mg/mL, ([IFN] stock=9.1 mg/mL). Buffers used were 20 mM sodium phosphate at pH 6.0 and 7.2; 20 mM NaAc at pH 3.0, 4.0, 4.5, 5.0 and 5.5; and 20 mM Glycine at pH 2.0 [0293]
  • The secondary structure content was estimated with several fitting programs, each of which decomposes the CD curve into four major structural components: α-helix, β-sheet, turns, and random coil. Two of those programs were provided with the CD instrument as an analysis facility. The first program uses seven reference proteins: Myoglobin, Lysozyme, Papain, Cytochrome C, Hemoglobin, Ribonuclease A and Chymotrypsin. The second uses Yang.REF reference file. [0294]
  • A third program, CCAFAST, uses the Convex Constraint Algorithm and is described in “Analysis of Circular Dichroism Spectrum of Proteins Using the Convex Constraint Algorithm: A Practical Guide”. (A. Perczel, K. Park and G. D. Fasman (1992) [0295] Anal. Biochem. 203: 83-93).
  • Deconvolution of the far UV scans over a range of pH volumes (2.0-7.2) indicates significant compaction of the secondary structure at pH 3.5. The near UV scan indicates a disruption of tertiary structure packing, and the far UV scan indicates that there is still significant secondary structure at this pH. [0296]
  • Results are illustrated in FIG. 8. [0297]
  • EXAMPLE 9 DSC of Insulin and Increasing Concentrations of GuHCl
  • DSC was performed with 6 mg/mL insulin (0.83 mM assuming a molecular weight of 6,000) in 50 mM phosphate buffer, pH 7.5. Each subsequent thermogram was corrected by background subtraction of a 0.6M guanidine-phosphate buffer solution. [0298]
  • Insulin was freshly prepared as a concentrated stock solution in 50 mM phosphate buffer, pH 7.5, and an appropriate aliquot was diluted in buffer, filtered though a 2 micron PTFE filter, and degassed for at least 20 minutes. The reference cell contained degassed buffer. [0299]
  • Scanning calorimetry was performed using 5 mg 0.83 mM porcine insulin (MW 6,000) per mL in 50 mM phosphate buffer, pH 7.5. All thermograms were performed on a Microcal MC-2 scanning calorimeter equipped with the DA2 data acquisition system operated in the upscale mode at 1° C./min (up to 90° C.), and data points were collected at 20 second intervals. All scans were initiated at least 20 degrees below the observed transitions for the active agent. All thermograms were corrected for baseline subtraction and normalized for the concentration of macromolecule. According to the methods of the Johns Hopkins Biocalorimetry Center, See, for example, Ramsay et al. [0300] Biochemistry (1990) 29:8677-8693; Schon et al. Biochemistry (1989) 28:5019-5024 (1990) 29: 781-788. The DSC data analysis software is based on the statistical mechanical deconvolution of a thermally induced macromolecular melting profile.
  • The effect of GuHCl on structure was assessed in DSC experiments where individual solutions were prepared in phosphate buffer, pH 7.5, containing denaturant diluted from a 5M stock solution to concentrations ranging for 0.5-2M. [0301]
  • Results are illustrated in Table 2 below. [0302]
    TABLE 2
    DSC of Insulin and Increasing Concentrations
    of Guanidine Hydrochloride
    Tm (Cp, max)
    (° C.)
    Insulin 0.0 M GuHCI 78.3
    Insulin + 0.5 M GuHCI 79.3
    Insulin + 1.0 M GuHCI 77.5
    Insulin + 2.0 M GuHCI 69.7
    Insulin + 3.0 M GuHCI no transition observed
  • EXAMPLE 10 Effect of Ionic Strength on the DSC Spectrum of Insulin
  • A sample containing 6 mg/mL insulin (0.83 mM in 50 mM phosphate buffer, pH 7.5, containing 0.25, 0.5, or 1.0M NaCl) was used. Thermograms were performed according to the procedure in Example 9 and were corrected by subtraction of a 0.5M NaCl-phosphate buffer blank as described above. [0303]
  • The effect of increasing ionic strength on structure was assessed in DSC experiments where individual solutions were prepared so as to contain NaCl at concentrations ranging from 0.25-3M. [0304]
  • Results are illustrated in Table 3 below. [0305]
    TABLE 3
    Effect of Ionic Strength on the DSC Spectrum
    of Insulin
    Tm (Cp, max)
    (° C.)
    Insulin 0.0 M NaCl 78.3
    Insulin + 0.25 M NaCl 80.7
    Insulin + 0.5 M NaCl 80.7
    Insulin + 1.0 M NaCl 80.7
  • EXAMPLE 10A Effect of Ionic Strength on the DSC Spectrum of rhGh
  • The method of Example 9 was followed substituting 5 mg/mL recombinant human growth hormone (rhGh) (225 μM based on M,22,128 of HGH) in 50 mM phosphate buffer, pH 7.5 containing either 0.5 or 1.0M NaCl, for the insulin. The thermograms were corrected by subtraction of a 0.5M NaCl-phosphate buffer blank. [0306]
  • Results are illustrated in Table 4 below. [0307]
    TABLE 4
    Effect of Ionic Strength on the DSC Spectrum of rhGh
    Tm (Cp, max) ΔH°
    (° C.) (kcal/mol)
    rhGh  0.0 M NaCl 75.2 191.0
    rhGh +  0.5 M NaCl 75.8 89.7
    rhGh + 10.0 M NaCl 76.5 50.5
  • EXAMPLE 11 Effect of pH on the DSC Spectrum of rhGH
  • 5 mg/mL rhGh were dissolved in buffer (0.17 mM in 50 mM phosphate buffer, assuming a molecular weight of 20,000). The pH of the solution was adjusted to the desired value, and all curves were corrected by baseline subtraction. [0308]
  • The effect of pH on structure was assessed by DSC according to the procedure of Example 9 where individual solutions were prepared in phosphate buffer ranging in pH value from 2.0 to 6.0. [0309]
  • Results are illustrated in Table 5 below. [0310]
    TABLE 5
    Effect of pH on the DSC Spectrum of rhGh
    Tm (Cp, max) ΔH°
    (° C.) (kcal/mol)
    pH 2.0 no transition observed no transition observed
    pH 3.0 no transition observed no transition observed
    pH 3.5 no transition observed no transition observed
    pH 4.0 ≈73.0 broad transition
    pH 5.0 75.0 161
    pH 6.0 75.2 191
    pH 7.5 (10 mg/mL) a) 73 (a) + (b) = 632
    b) 75
  • EXAMPLE 12 Effect of GuHCl on the DSC Spectrum of rhGh
  • An initial scan of rhGh was performed at 10 mg/mL in the absence of GuHCl (0.33 mM assuming 20,000 molecular weight). Subsequently, the concentration of rhGh was lowered to 5 mg/mL (0.17 mM) in 50 mM phosphate buffer, pH 7.5 containing varying concentrations of GuHCl. Each subsequent thermogram was corrected by background subtraction of a 0.5 M guanidine-phosphate buffer solution. The thermograms were corrected by subtraction of a 0.5M NaCl-phosphate buffer blank. Scans were performed according to the procedure of Example 9. [0311]
  • Results are illustrated in Table 6 below. [0312]
    TABLE 6
    Effect of Guanidine Hydrochloride on DSC Spectrum of rhGh
    DOMAIN A DOMAIN B
    Tm (Cp, max) Tm (Cp, max) ΔH°
    (° C.) (° C.) (kcal/mol)
    rhGh 72.6 74.3 632 
    rhGh + 0.5 M 71.5 not defined, 48
    GuHCl but present
    rhGh + 1.0 M GuHCl 70.9 absent 109 
    rhGh + 1.5 M GuHCl 69.7 absent 12
    rhGh + 2.0 M GuHCl 70.0 absent 58
    rhGh + 2.5 M GuHCl 70.7 absent 99
  • EXAMPLE 13 pH Dependence of α-interferon Conformation
  • α-interferon stock (9.1 mg/mL) was diluted with buffer to a concentration of 0.6 mg/mL. The sample was dialyzed overnight in buffer (volume ratio of α-interferon to buffer was 1:4000). Since there was no extinction coefficient provided, concentration of the sample used was determined by comparison of absorption spectra of the sample before and after dialysis. For each particular pH, the absorbance of the nondialyzed α-interferon of known concentration was measured at 280 nm. Then after dialysis, absorbance was read again to account for the protein loss, dilution, etc. Buffer conditions and α-interferon concentrations were: [0313]
  • pH 3.0: Buffer—20 mM NaAc. [IFN]=0.50 mg/mL; [0314]
  • pH 4.1: Buffer—20 mM NaAc. [IFN]=0.53 mg/mL; [0315]
  • pH 5.0: Buffer—20 mM NaAc. [IFN]=0.37 mg/mL; [0316]
  • pH 6.0: Buffer—20 mM Na Phosphate. [IFN]=0.37 mg/mL; [0317]
  • pH 7.2: Buffer—20 mM Na Phosphate. [IFN]=0.48 mg/mL. [0318]
  • DSC scans were performed according to the procedure of Example 9. Although clear, transparent solutions of α-interferon were obtained for every pH at room temperature, there were noticeable signs of precipitation at pH 5.0 and 6.0 after the temperature scans. [0319]
  • Results are illustrated in Table 7 below. [0320]
    TABLE 7
    α-Interferon - pH dependence DSC
    pH Tm° C. ΔH cal/mol
    7.2 66.84 732717
    6.0 65.34  45580
    5.0 67.32  69782
    4.1 65.64  60470
    3.0
  • EXAMPLE 14 Concentration Effect of GuHCl on α-Interferon Conformation
  • GuHCL/α-interferon samples were prepared according to the method of Example 4. DSC scans were performed according to the procedure of Example 9. [0321]
  • Results are illustrated in Table 8 below. [0322]
    TABLE 8
    α-Interferon in GuHCl DSC
    [GuHCl] M Tm ° C. ΔH cal/mol
    0.0 67.12 72562
    0.5 64.43 50827
    1.0 63.04 41705
    1.5 60.11 29520
    2.0 56.32 24980
    3.0 45.90 20577
    4.0
  • Examples 3-14 illustrate that ionic strength, guanidine hydrochloride concentration, and pH result in changes in the Tm of active agents, indicating a change in conformation. This was confirmed by fluorescence spectroscopy. The reversible intermediate conformational states can be used as templates to prepare mimetics. [0323]
  • EXAMPLE 15 Preparation of α-Interferon Intermediate State Mimetics
  • An intermediate conformational state of α-interferon is determined. A peptide mimetic having the secondary and tertiary structure of the intermediate state is prepared. [0324]
  • EXAMPLE 16 Preparation of Insulin Intermediate State Mimetics
  • The method of Example 15 is followed substituting an insulin for the α-interferon. [0325]
  • EXAMPLE 17 Preparation of rhGh Intermediate State Mimetics
  • The method of Example 15 is followed substituting recombinant human growth hormone for the α-interferon. [0326]
  • EXAMPLE 18 Titration of α-interferon as Measured by Intrinsic Tryptophan Fluorescence
  • A stock solution of 9.1 mg/mL α-interferon in 20 mM sodium phosphate buffer at pH 7.2 was prepared. A stock solution of perturbant was prepared by dissolving 800 mg of perturbant (L-arginine acylated with cyclohexanoyl chloride) in 2 mL of 20 mM Sodium Phosphate buffer (pH7). [0327]
  • Samples were prepared by diluting the α-interferon with the sodium phosphate buffer and perturbant stock solution at various perturbant concentrations. Diluted samples were allowed to come to equilibrium by incubation for approximately 30 minutes at room temperature prior to measurement. [0328]
  • Fluorescence from the endogenous tryptophan resident of α-interferon were measured according to the procedure of Example 3. The perturbant did not contain a fluoophore. [0329]
  • Results are illustrated in FIG. 9. [0330]
  • EXAMPLE 19 Differential Scanning Colorimetry of α-interferon and Perturbant
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant was weighed out to make perturbant stock solutions. α-interferon stock was diluted in the buffer. α-interferon solution was not dialyzed prior to experiments for the purpose of having the same active concentration for the whole set. [0331]
  • DSC thermograms were generated with α-interferon at a concentration of 0.64 mg/ml and a perturbant (phenylsulfonyl-para-aminobenzoic acid purified to >98% (as determined by reverse phase chromatography prior to generation of the spectra)) at perturbant concentrations of 5, 10, 25 and 100 mg/ml. DSC was conducted on a DASM-4 differential scanning calorimeter interfaced to an IBM PC for automatic collection of the data. The scanning rate was 60° C./h. [0332]
  • Results are illustrated in Table 9 below and FIG. 10. [0333]
  • COMPARATIVE EXAMPLE 19* Different Scanning Calorimetry of α-interferon
  • The method of Example 19 was followed substituting α-interferon without perturbant. Results are illustrated in Table 9 below and FIG. 10. [0334]
    TABLE 9
    α-Interferon + Perturbant DSC
    Perturbant - mg/ml Tm ° C. ΔH cal/mol
    0 67.12 72562  
    5 64.37 60151  
    10 62.3 53161  
    25 58.15 35393  
    100 46.18  5439.3
  • DSC scans where the added concentration of perturbant ranged from 0-100 mg/mL show induced conformational changes in the α-interferon that occur in a concentration dependent manner. At 100 mg/mL of the perturbant, the thermogram indicated that the α-interferon Cp vs. Tm curve was a flat line. The flat Cp vs. Tm curve obtained at 100 mg/mL of perturbant indicates that hydrophobic residues within the α-interferon molecule became solvent exposed. It is clear that the perturbant was able to change the structure of α-interferon in a concentration dependent manner. [0335]
  • EXAMPLE 20 Dialysis Experiments—Reversibility of Complexing with the Perturbant
  • An α-interferon stock solution at a concentration of 9.1 mg/mL was diluted with buffer to an α-interferon concentration of 0.6 mg/mL. DSC was performed according to the procedure of Example 19. [0336]
  • Results are illustrated in FIG. 11A. [0337]
  • α-interferon (0.6 mg/ml) and the perturbant of Example 19 (100 mg/ml) were mixed with no apparent changes in the Cp of the solution. This solution was then dialyzed overnight into phosphate buffer, and the thermogram was rerun. Results are illustrated in FIG. 11B. [0338]
  • The dialyzed sample had essentially the same Tm and the same area under the Cp vs. Tm curve as it did prior to addition of the perturbant. This indicated that not only was the perturbant able to induce conformational changes in the protein but that this process was reversible. Dilution was enough of a driving force to effect disengagement of the perturbant from the active agent. [0339]
  • EXAMPLE 21 Perturbant and α-Interferon DSC
  • The method of Example 4 was followed, substituting the perturbant of Example 19 for the GuHCl. [0340]
  • Results are illustrated in FIG. 12. [0341]
  • The DSC experiments on the equilibrium denaturation of α-interferon indicate the existence of intermediate conformations of the molecule. ΔH vs. Tm plots indicate the energetics of intermediate conformations occupied by α-interferon at each set of experimental conditions. [0342]
  • EXAMPLE 22 Perturbant Concentration Dependent Change in α-interferon
  • The method of Example 19 was followed substituting cyclohexanoyl phenylglycine for the perturbant. [0343]
  • Results are illustrated in Table 10 below and in FIG. 13. [0344]
    TABLE 10
    α-Interferon + Perturbant DSC
    Perturbant mg/ml Tm ° C. ΔH cal/mol
     0 67.12 72562
     5 63.00 42299
    10 59.49 43058
    25 52.79 27237
    100    0
  • Cyclohexanoyl phenylglycine induced conformational changes in α-interferon that were concentration dependent. [0345]
  • EXAMPLE 23 Perturbant and α-Interferon DSC
  • The method of Example 4 was followed substituting the perturbant of Example 30 for the GuHCl. [0346]
  • Results are illustrated in FIG. 14. [0347]
  • The ΔH v. Tm plot indicates the existence of an equilibrium intermediate conformation of α-interferon that is stable at below 5 and 25 mg/ml of added cyclohexanoyl phenylglycine perturbant. [0348]
  • EXAMPLE 24 Isothermal Titration Calorimetry of α-Interferon with Perturbant
  • Isothermal titration calorimetry of perturbant complexing with α-interferon was performed at 25° C. at two different pH's. The buffers used were 20 mM NaPhosphate for pH 7.2 and 20 mM NaAc for pH 3.0. α-interferon solution was dialyzed before the experiment to reach the appropriate pH. Dry perturbants were weighed and diluted in dialysate. [0349]
  • ITC was conducted on a MicroCal OMEGA titration calorimeter (MicroCal Inc.—Northampton, Mass.). Data points were collected every 2 seconds, without subsequent filtering. α-interferon solution placed in 1.3625 mL cell was titrated using a 250 μL syringe filled with concentrated perturbant solution. A certain amount of titrant was injected every 3-5 minutes for up to 55 injections. [0350]
  • A reference experiment to correct for the heat of mixing of two solutions, was performed identically except that the reaction cell was filled with buffer without active agent. [0351]
  • Analysis of the data was performed using the software developed at the Johns Hopkins University Biocalorimetry Center. [0352]
  • The titration at pH 7.2 included 53 injections of 2 μL of the perturbant of Example 30 (50 mg/mL=191.6 mM (MW 261)) and α-interferon (1.3 mg/mL=0.067 mM (MW 19400)). [0353]
  • Results are illustrated in FIG. 15. [0354]
  • Curve fitting indicated multiple independent sites: [0355]
  • n (1)=121.0354 where n=# of completed perturbant molecules [0356]
  • ΔH (1)=58.5932 cal/Mole perturbant [0357]
  • log 10 Ka (1)=2.524834 where Ka=association constant [0358]
  • x-axis units are concentration of carrier in mM. [0359]
  • y-axis units represent heat/injection expressed in calories. [0360]
  • At [0361] pH 3, complexing resulted in a negative enthalpy.
  • COMPARATIVE EXAMPLE 24* Isothermal Titration Calorimetry of Perturbant
  • The method of Example 24 was followed, substituting 53 injections of 2 μl for the perturbant (50 mg/mL=191.6 mM) [IFN]=0 mg of Example 22 without active agent. [0362]
  • Example 24 and Comparative Example 24* illustrate that α-interferon has a positive enthalpy and a binding constant (K[0363] d≈10−3M).
  • EXAMPLE 25 Isothermal Titration Calorimetry of α-interferon and Perturbant Complexing
  • The method of Example 24 was followed substituting the perturbant of Example 19 for the perturbant of Example 22. [0364]
  • The titration at pH 7.2 included two runs of 55 injections each of 5 μL of perturbant (50 mg/mL=181 mM (FW 277)) and α-interferon (2.31 mg/mL=0.119 mM, (MW 19400)). [0365]
  • Results are illustrated in FIG. 16. [0366]
  • Curve fitting indicated multiple independent sites: [0367]
  • n (1)=55.11848 where n=# of complex perturbant molecules [0368]
  • ΔH (1)=−114.587 cal/Mole perturbant [0369]
  • log 10 Ka (1)=2.819748 where Ka=association constant [0370]
  • x-axis units are concentration of carrier in mM. [0371]
  • y-axis units represent heat/injection expressed in calories. [0372]
  • Complexing of perturbant to α-interferon at pH 3.0 resulted in precipitation of the complex out of the solution. Due to the heat effect produced by this process, it was impossible to measure the complexing parameters. [0373]
  • COMPARATIVE EXAMPLE 25* Isothermal Titration Calorimetry of Perturbant
  • The method of Example 25 was followed, substituting 55 injections of 5 μl of the perturbant of Example 26(50 mg/mL=181 mM) in 20 mM sodium phosphate pH 7.2 without active agent. [0374]
  • The perturbant of Example 19 complexed with α-interferon resulted in a negative enthalpy and a comparable binding constant to that of the perturbant of Example 22 and α-interferon. [0375]
  • Examples 24 and 25 indicate that the stronger the perturbant complexes with the active agent and the more thermodynamically stable the intermediate state of the active agent, the greater the bioavailability of the active agent. [0376]
  • Therefore, by plotting the ΔH v. Tm curve for an active agent and a perturbant, those perturbants that induce little or no enthalpic change over the broadest range of Tm would be preferred perturbants. It is believed that perturbants that stabilize the intermediate state to a greater extent will result in more efficient delivery of the active agent. [0377]
  • EXAMPLE 26 Comparison of the Effects of Three Perturbants on ΔH vs. Tm Plots with α-interferon
  • DSC experiments were carried out according to the procedure of Example 19, with 0.5 mg/ml α-interferon mixed with (1) benzoyl para-amino phenylbutyric acid, (2) the perturbant of Example 22, or (3) the perturbant of Example 19. [0378]
  • Benzoyl para-amino phenylbutyric acid was poorly soluble under the buffer conditions. Maximum concentration at which the solution was still transparent at room temperature was ˜8 mg/mL. Therefore, the concentrations of the perturbant used were 2, 4, and 6 mg/mL. Results are illustrate in FIGS. 17 and 18. The dashed line in FIG. 17 represents the linear least squares, and the regression equation is at the top of FIG. 17. [0379]
  • Y=−1.424e 5+3148.8x
  • R=0.9912
  • FIGS. 17 and 18 illustrate that conformational changes in α-interferon are more readily produced by benzoyl para-amino phenylbutyric acid than by the perturbants of Examples 22 and 19, and that such changes are more readily produced by the perturbant of Example 22 than by the perturbant of Example 19. [0380]
  • EXAMPLE 27 Isothermal Titration Calorimetry of α-Interferon and Complexing
  • ITC was performed according to the method of Example 24 with 40 injections of 5 μL of the perturbant benzoyl para-amino phenylbutyric acid (7.5 mg/mL=24.59 mM, (FW 305)) and α-interferon (2.5 mg/mL=0.129 mM, (MW 19400)). [0381]
  • Results are illustrated in FIG. 19. [0382]
  • Curve fitting indicated multiple independent sites: [0383]
  • n (1)=23.69578 where n=# of complexed perturbant molecules [0384]
  • ΔH (1)=791.5726 cal/Mole perturbant [0385]
  • log 10 Ka (1)=3.343261 where Ka=association constant [0386]
  • x-axis represents concentration of carrier in mM. [0387]
  • y-axis represents heat/injection expressed in calories. [0388]
  • COMPARATIVE EXAMPLE 27*
  • ITC was performed according to the method of Example 27 with 40 injections of 5 μl of the perturbant benzoyl para-amino phenylbutyric acid (7.5 mg/mL=24.59 mM) in 20 mM NaPhosphate pH 7.2 buffer, without active agent. [0389]
  • The apparent dissociation constant for the perturbant of Example 26 is greater than that for the perturbant of Example 30 (10[0390] −4M) at pH 7.
  • Therefore, benzoyl para-amino phenylbutyric acid complexes more strongly to α-interferon and induces the native state-reversible intermediate conformational state at lower concentrations of perturbant. [0391]
  • EXAMPLES 28-30 Isothermal Titration Calorimetry of rhGH at pH 7.5 and 4.0 with Different Perturbants
  • The ability of rhGh to complex with various perturbants was assessed by ITC using a Microcal Omega titrator, usually equilibrated at 30° C. The sample cell of the calorimeter was filled with degassed rhGH (usually at 0.25 mM) prepared in 50 mM phosphate buffer, pH 7.5 or 4.0. The perturbant (cyclohexanoyl chloride modified L-tyrosin (a), salicyloyl modified L-phenylalanine (b), or phenylsulfonyl-para-amino benzoic acid (c)) was then placed in the dropping syringe at 1 mM (for pH 7.5) and 2.5 mM, (for pH 4.0). Twenty to twenty-five 10 μl injections were made into rapidly mixing (400 rpm) solution with 2 minute intervals between injections. [0392]
  • Initial concentration of perturbant placed in the calorimeter sample cell assumed a formula weight of 200 for each perturbant. The pH of each solution was checked after dissolution, but no adjustments of the pH were required. All experiments were performed at 30° C. Initial concentration of rhGh placed in the dropping syringe assumed a molecular weight of 20,000 for rhGh. The pH of each solution was checked after dissolution, but no adjustment of the pH was required. [0393]
  • The heats of reaction were determined by integration of the observed peaks. To correct for heat of mixing and dilution, a control experiment was also performed under identical conditions where aliquots of the test perturbant or rhGh were added to buffer solution only. The sum total of the heat evolved was plotted against the total perturbant concentration to produce the isotherm from which the association constant (K[0394] A, M), enthalpy change (ΔH, kcal/mol), entropy change (ΔS (eu), and N, and the stoichiometry of perturbant molecules complexed per equivalent of complexed supramolecular complex, were determined by curve-fitting the binding isotherm against the binding equation described for perturbant complexing in a supramolecular complex possessing one set of independent perturbant complexing sites. The data were deconvoluted using the nonlinear least squares algorithm supplied in the software of the manufacturer.
  • Results are illustrated in Table 11 below. [0395]
    TABLE 11
    Isothermal Titration Calorimetry of rhGH at pH
    7.5 and 4.0 with Different Perturbants
    rhGh KD ΔH ΔS
    Perturbant (mM) (M) (kcal/mol) (eu) N
    pH 7.5
    A at 0.25 mM 1.0 9.88 × 10−5 +1.4 +23.5 7.0
    B at 0.25 mM 1.0 1.11 × 10−6 +2.1 +35.0 0.7
    C at 0.25 mM 1.0 1.11 × 10−9 +0.8 +44.0 10.0
    pH 4.0
    A at 0.25 mM 1.0 7.81 × 10−5 −1.5 −5.6 2.3
    B at 0.25 mM 1.0 1.61 × 10−9 −35.6 −90.0 155.9
    C at 0.25 mM 1.0 2.67 × 10−8 −1.2 −30.0 122.0
  • The positive ΔS values at pH 7.5 indicate that complexing at this pH results in structural change. [0396]
  • EXAMPLES 31 AND 32 Pancreatin Inhibition Assay with α-lnterferon and Perturbants
  • The assay for pancreatin activity was prepared as follows: 0.1 mL of a stock solution of α-interferon (9.1 mg/mL, 20 mM NaH[0397] 2PO4, pH 7.2) (Schering-Plough Corp.) was added to 2.5 mL of either phenylsulfonyl-para-aminobenzoic acid perturbant (46) or cyclohexanoyl phenylglycine perturbant (47) (200 mg/mL) in 5 mM KH2PO4, pH 7.0. Incubation was carried out at 37° C. for 30 and 60 minutes following the addition of 0.1 mL of USP pancreatin (20 mg/mL) (Sigma Chemical Co.) 0.1 mL aliquots were withdrawn at those times points. Enzyme reactions were stopped by the addition of protease inhibitors (Aprotinin and Bowman-Birk Inhibitor (BBI), each at 2 mg/mL) and were diluted five-fold to quantitate α-interferon left intact. A reverse phase HPLC method using a Butyl C-4 cartridge (3.0×46 cm, Rainin) and employing gradient elution between 0.1% TFA/water and 90% ACN in 0.1% TFA coupled with UV detection at 220 nm was used for separating and quantitating α-interferon. The α-interferon at 0 minutes was quantitated from an aliquot prior to the addition of pancreatin and was taken to be 100%.
  • Results are illustrated in FIG. 20. [0398]
  • Examples 31 and 32 illustrate that both supramolecular complexes resisted enzymatic degradation. However, in additional testing no correlation was shown between the enzyme inhibitors potency and the ability to deliver drug. [0399]
  • EXAMPLE 33 DSC of Heparin at pH 5.0
  • DSC thermograms of heparin at pH 5.0 were conducted according to the method of Example 9 using pH, GuHCl, and ionic strength as perturbants. [0400]
  • Thermograms were corrected by subtraction of a heparin 0.05M NaCl-phosphate buffer blank, but an individual blank was not used for each NaCl concentration. [0401]
  • Results are illustrated in Tables 12-14 below and in FIG. 21. [0402]
    TABLE 12
    Effects of pH on the DSC Spectrum of 20 μg/ml Heparin
    in 50 mM Phosphate Buffer
    ΔH ΔHvH
    Tm (Cp, max) (kcal/mol) (kcal/mol)
    pH 6.0 62.5 232.1 13.8
    pH 6.5 (a) 62.7 213.9
    (b) 71.8 751.9 56.8
    pH 7.0 (a) 47.1 187.1
    (b) 72.9 136.4 27.6
    pH 7.5 66.2 499.4 83.8
  • [0403]
    TABLE 13
    Effects of 10M Guanidine Hydrochloride in 50 mM Phosphate Buffer
    on the DSC Spectrum of Heparin
    ΔH ΔHvH
    Tm (Cp, max) (kcal/mol) (kcal/mol)
    heparin 67.2 499.4 83.8
    heparin + 0.5M GuHCl 50.5 287.3 170.9
    heparin + 1.0M GuHCl 60.5 415.0 97.1
    heparin + 1.5M GuHCl 1716.5 24.3
    heparin + 2.0M GuHCl 2533.7 19.2
  • [0404]
    TABLE 14
    Effect of Ionic Strength on the DSC Spectrum of 20 μg/ml
    of Heparin in 50 mM Phosphate Buffer pH 7.0
    ΔH ΔHvH
    Tm (Cp, max) (kcal/mol) (kcal/mol)
     0.0M NaCl 47.1 187.1 72.9 136.4
    0.25M NaCl 46.1 0.112 not present
    0.50M NaCl 41.6 0.094 not present
    0.75M NaCl 27.5 0.00 not present
     1.0M NaCl no transition observed
  • These data indicate that non-proteinaceous active agents are able to change conformation in response to a perturbant. [0405]
  • EXAMPLE 34 Column Chromatography of Heparin and Perturbants
  • The following materials were used: [0406]
  • Column: [0407]
  • 10 mm×30 cm, low pressure, glass column from Pharmacia w/adjustable bed volume. The bed volume used was 22 cm at a pressure of 0.8 Mpa. [0408]
  • Packing: [0409]
  • Heparin covalently bonded to Sepharose CL-6B with no linker molecule. [0410]
  • Sepharose fractionation range: 10,000-4,000,000. [0411]
  • The density of heparin was 2 mg/cc as per Pharmacia Q.C. Department. [0412]
  • Conditions: [0413]
  • The mobil phase was 67 mM phosphate buffer, pH7.4. [0414]
  • The flow rate was 1.5 mL/min isocratic. [0415]
  • The run time was 45 minutes. [0416]
  • Sample detection was done with a Perkin Elmer refractive index detector. [0417]
  • Column integrity was confirmed by injecting protamine and observing a retention time greater than 1 hour. Void volume was determined by injecting water and measuring time of elution. [0418]
  • Each of the perturbants of Table 15 below (5 mg) was independently dissolved in 1 mL of mobil phase and injected (100 ul) into the column. Time of elution was measured. K′ value was determined by using the following equation (as per USP): [0419]
  • K′=(Ret. time Carrier/Ret. time Water)−1
  • The results were compared between each perturbant as well as their respective in vivo performance in FIG. 29. K′ (the degree of retardation) values in the figure have been corrected by subtraction of the K′ value determined from the sepharose column from the K′ value determined from the heparin-sepharose column. [0420]
    TABLE 15
    PERTURBANTS
    cyclohexylidenebutyric acid(2)-Na salt # 1
    cylcohexanebutyroyl(2-)aminobutyric acid(4) #2
    phenylacetyl-para-aminobutyric acid # 3
    ortho-methylcyclohexanoyl-aminobutyric acid(4) #4
    phenylacetyl-aminohexanoic acid(6-) #5
    cinnamoyl-para-aminophenylbutyric acid # 6
    cyclohexanebutyroyl(2-)-para-aminophenylbutyric acid #7
    hydrocinnamoyl-para-aminophenylbutyric acid # 8
    cyclohexanebutyroyl(2-)-leu-leu #9
    cyclohexanebutyroyl(2-)-gly # 10 
  • EXAMPLE 35 Comparison of the Effects of Six Perturbants on ΔH vs. Tm Plots with DPPC
  • DSC experiments were carried out according to the procedure of Example 19, with 1.0 mg/ml dipalmitoylphosphatidylcholine (DPPC) mixed with perturbants XI, L, LII, LIII, and LIV. The concentrations of the perturbant were varied from 0 to 20 mg/ml. [0421]
  • Results are illustrated in FIG. 22. [0422]
  • EXAMPLE 36 Differential Scanning Colorimetry of DPPC and Perturbant Compound L
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant L was weighed out to make perturbant stock solutions. DPPC stock solution was prepared in the buffer. [0423]
  • DSC thermograms were generated with DPPC at a concentration of 1.0 mg/ml. The perturbant concentrations used were 0, 5, 10, and 20 mg/ml. The DSC was performed as described in Example 19. [0424]
  • Results are illustrated in FIG. 23. [0425]
  • EXAMPLE 37 Differential Scanning Colorimetry of DPPC Perturbant Compound L and rhGH
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant L was weighed out to make perturbant stock solutions. DPPC stock solution was prepared in the buffer. rhGH solution was prepared as described in Example 10A. [0426]
  • DSC thermograms were generated with DPPC at a concentration of 1.0 mg/ml. Samples having DPPC alone; DPPC with 10 mg/ml of perturbant; DPPC with 0.3 mg/ml of rhGH; and DPPC, 10 mg/ml of perturbant and 0.3 mg/ml of rhGH were prepared and analyzed. The DSC was performed as described in Example 19. [0427]
  • Results are illustrated in FIG. 24. [0428]
  • EXAMPLE 38 Differential Scanning Colorimetry of DPPC Perturbant Compound LII and rhGH
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant LII was weighed out to make perturbant stock solutions. DPPC stock solution was prepared in the buffer. rhGH solution was prepared as described in Example 10A. [0429]
  • DSC thermograms were generated with DPPC at a concentration of 1.0 mg/ml. Samples having DPPC alone; DPPC with 5 mg/ml of perturbant; DPPC with 0.3 mg/ml of rhGH; and DPPC, 5 mg/ml of perturbant and 0.3 mg/ml of rhGH were prepared and analyzed. The DSC was performed as described in Example 19. [0430]
  • Results are illustrated in FIG. 25. [0431]
  • EXAMPLE 39 Differential Scanning Colorimetry of DPPC Perturbant Compound XI and rhGH
  • Perturbant binding DSC was conducted using 20 mM NaPhosphate buffer at pH 7.2. Dry perturbant XI was weighed out to make perturbant stock solutions. DPPC stock solution was prepared in the buffer. rhGH solution was prepared as described in Example 10A. [0432]
  • DSC thermograms were generated with DPPC at a concentration of 1.0 mg/ml. Samples having DPPC alone; DPPC with 2 mg/ml of perturbant; DPPC with 0.3 mg/ml of rhGH; and DPPC, 2 mg/ml of perturbant and 0.3 mg/ml of rhGH were prepared and analyzed. The DSC was performed as described in Example 19. [0433]
  • Results are illustrated in FIG. 26. [0434]
  • EXAMPLE 40 Dynamic Light Scattering of Compound L
  • Solutions of compound L were prepared in a 10 mM phosphate buffer at a pH of 7.0. The concentrations tested were 10, 15, and 20 mg/ml. The solutions were analyzed using standard microscopic light scattering techniques. [0435]
  • Results are illustrated in FIG. 27. [0436]
  • EXAMPLES 41 AND 42 In Vivo Evaluation of Recombinant Growth Hormone in Rats
  • Dosing compositions were prepared by mixing the modified amino acids and recombinant human growth hormone (rhGH) as listed in Table 16 below in a phosphate buffer solution at a pH of about 7-8. [0437]
  • Rats were administered the dosing composition by sublingual or intranasal administration. Delivery was evaluated by using an ELISA assay for rhGH from Medix Biotech, Inc. For intracolonic administration, a sample was prepared and dosed to fasted rats at 25 mg/kg of carrier in a buffered solution containing propylene glycol (0-50%) and 1 mg/kg rhGH. [0438]
  • Results are illustrated in Table 16 below. [0439]
  • COMPARATIVE EXAMPLE 41A
  • rhGH (6 mg/ml) was administered by oral gavage to a rat, and delivery was evaluated according to the procedure of Example 41A. [0440]
  • Results are illustrated in Table 16 below. [0441]
    TABLE 16
    In Vivo Delivery of rhGH
    Carrier Drug Method of Mean Peak Serum
    Dose Dose Admin- Levels of rhGH
    Example Carrier (mg/kg) (mg/kg) istration (ng/mL)
    42A none  0 6 oral <10 +/− 10  
    41 XIX-1 100  3 sublingual 119.14 +/− 65.6 
    42 XIX-1 25 1 intranasal 92.7 +/− 73.2
  • EXAMPLES 43-54 In Vivo Evaluation of Recombinant Growth Hormone in Rats
  • Preparation of Dosing solutions [0442]
  • The delivery agents were reconstituted with distilled water and adjusted to pH 7.2-8.0 with either aqueous hydrochloric acid or aqueous sodium hydroxide. A stock solution of rhGH was prepared by mixing rhGH, D-mannitol and glycine and dissolving this mixture in 2% glycerol/water. The stock solution was then added to the delivery agent solution. Several delivery agent to active agent ratios were studied. [0443]
  • In vivo experiments [0444]
  • Male Sprague-Dawley rats weighing 200-250 g were fasted for 24 hours and administered ketamine (44 mg/kg) and chlorpromazine (1.5 mg/kg) 15 minutes prior to dosing. The rats were administered one of the dosing solutions described above by subcutaneous injection, intranasal instillation, or sublingual instillation. Blood samples were collected serially from the tail artery for serum calcium concentration determination or serum rhGH concentrations. The dose of rhGH administered in these experiments was 0.1 mg/kg. [0445]
  • Serum rhGH concentrations were quantified by an rhGH enzyme immunoassay test kit. The results are given in Table 17 and FIGS. 28 and 29. [0446]
  • In FIG. 29 the circles represent the response following SL dosing of an aqueous solution of compound CXXIII-H and rhGH. The squares represent the response following IN dosing of an aqueous solution of compound CXXIII-H and rhGH. The triangles represent the response following IC dosing of an aqueous solution of compound CXXIII-H and rhGH. The dose of compound CXXIII-H was 25 mg/kg and the dose of rhGH was 1 mg/kg. [0447]
  • COMPARATIVE EXAMPLE 43A
  • rhGH (1 mg/kg) was administered by oral gavage to a rat, and delivery was evaluated according to the procedure of Example 43. [0448]
  • Results are illustrated in Table 17 below. [0449]
    TABLE 17
    Delivery Agent Enhancement of Recombinant Human Growth Hormone
    (rhGH) Bioavailability Administered by Subcutaneous Administration.
    Delivery Agent Dose Peak Serum [rhGH]
    Example Deliver Agent (mg/kg) (ng/mL)
    43 CXXIII-H 1.0 22 ± 3
    43A None 0.0  4 ± 2
    44 CXXIII-H 2.5 25 ± 5
    45 CXXIII-H 25 30 ± 6
    46 CXI-G 2.5 16 ± 2
    47 LVIII-A 1.0 29 ± 10
    48 LXXXVI-B 1.0 22 ± 7
    49 LXXXVI-B 2.5 23 ± 5
    50 LXI-A 2.5 26 ± 5
    51 CX-1 1.0 15 ± 3
    52 CXV-G 1.0 25 ± 3
    53 LXVI-A 1.0 33 ± 5
    54 CIX-1 1.0 16 ± 3
  • EXAMPLES 55-60 In Vivo Evaluation of Salmon Calcitonin in Rats
  • Preparation of Dosing solution [0450]
  • The delivery agents were reconstituted with distilled water and adjusted to pH 7.2-8.0 with either aqueous hydrochloric acid or aqueous sodium hydroxide. A stock solution of sCT was prepared by dissolving sCT in citric acid (0.085N). The stock solution was then added to the delivery agent solution. Several different delivery agent to active agent ratios were studied. [0451]
  • In vivo experiments [0452]
  • Male Sprague-Dawley rats weighing 200-250 g were fasted for 24 hours and administered ketamine (44 mg/kg) and chlorpromazine (1.5 mg/kg) 15 minutes prior to dosing. The rats were administered one of the dosing solutions described above by subcutaneous injection. Blood samples were collected serially from the tail artery for serum calcium concentration. [0453]
  • Serum calcium concentrations were quantified by the o-cresolphthalein complexone method (Sigma) using a UV/VIS spectrophotometer (Perkin Elmer). The results are given in Table 5. [0454]
  • EXAMPLES 55A
  • Salmon calcitonin was administered by oral gavage to rats, and delivery was evaluated according to the Procedure of Example 55. The results are given in Table 18 below. [0455]
    TABLE 18
    Delivery Agent Enhancement of Salmon Calcitonin (sCT, dosed
    at 0.2 μg/kg) Bioavailability Administered by Subcutaneous
    Administration.
    Delivery Agent Dose Percent Decrease in
    Example Deliver Ageny (μg/kg) Serum Calcium
    55 CXXIII-H 2 17 ± 3
    55A None 0 17 ± 2
    56 CXXIII-H 20 25 ± 4
    57 CXXIII-H 200 25 ± 5
    58 CXXIII-H 2000 26 ± 5
    59 CXI-G 20 21 ± 4
    60 CXIV-G 20 20 ± 3
  • All patents, applications, test methods, and publications mentioned herein are hereby incorporated by reference. [0456]
  • Many variations of the present invention will suggest themselves to those skilled in the art in light of the above detailed disclosure. All such modifications are within the full extended scope of the appended claims. [0457]

Claims (111)

In the claims:
1. A method for delivering, by the subcutaneous route, a biologically active agent to a subject in need of said biologically active agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a complexing perturbant to reversibility transform said biologically active agent to said intermediate state and to form a subcutaneously deliverable supramolecular complex,
said perturbant having a molecular weight between about 150 to about 600 daltons, and having at least one hydrophilic moiety and at least one hydrophobic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent,
said biologically active agent not forming a microsphere with said perturbant, and
said perturbant being present in an amount effective for subcutaneous delivery of said biologically active agent; and
(c) subcutaneously administering said supramolecular complex to said subject.
2. A method as defined in
claim 1
, further comprising
(d) after said administering step, removing said perturbant from said supramolecular complex to transform said biologically active agent to said native state.
3. A method as defined in
claim 2
, wherein step (d) comprises diluting said supramolecular complex.
4. A method as defined in
claim 1
, wherein said intermediate state has a ΔG ranging from about −20 kcal/mole to about 20 kcal/moles relative to said native state.
5. A method as defined in
claim 1
, wherein said biologically active agent is selected from the group consisting of a peptide, a mucopolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
6. A method as defined in
claim 5
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
7. A method as defined in
claim 1
, wherein said perturbant comprises a proteinoid.
8. A method as defined in
claim 1
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
9. A method as defined in
claim 1
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
10. A method as defined in
claim 1
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
11. A method as defined in
claim 1
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
12. A method as defined in
claim 1
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2to C10 alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
13. A method for preparing a subcutaneously deliverable biologically active agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states; and
(b) exposing said biologically active agent to a complexing perturbant to reversibility transform said biologically active agent to said intermediate state and to form a subcutaneously deliverable supramolecular complex,
said perturbant having a molecular weight ranging from about 150 to about 600 daltons, and having at least one hydrophilic moiety and at least one hydrophobic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent;
said biologically active agent not forming a microsphere with said perturbant; and
said perturbant being present in an amount effective for subcutaneous delivery of said biologically active agent.
14. A method as defined in
claim 13
, wherein said intermediate state has ΔG ranging from about −20 kcal/mole to about 20 kcal/moles relative to said native state.
15. A method as defined in
claim 13
, wherein said biologically active agent is selected from the group consisting of a peptide, a micropolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
16. A method as defined in
claim 15
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
17. A method as defined in
claim 13
, wherein said perturbant comprises a proteinoid.
18. A method as defined in
claim 13
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
19. A method as defined in
claim 13
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
20. A method as defined in
claim 13
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
21. A method as defined in
claim 13
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
22. A method as defined in
claim 13
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2to C10 alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
23. A subcutaneous delivery composition comprising a supramolecular complex comprising:
(a) a biologically active agent in an intermediate conformational state non-covalently complexed with
(b) a complexing perturbant having a molecular weight ranging from about 150 to about 600 and having at least one hydrophilic moiety and at least one hydrophobic moiety;
wherein said intermediate state is reversible to said native state and is conformationally between a native conformational and a denatured conformational state of said biologically active agent and said composition is not a microsphere; and said perturbant being present in an amount effective for subcutaneous delivery of said biologically active agent.
24. A composition as defined in
claim 23
, wherein said biologically active agent is selected from the group consisting of a peptide, a micropolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
25. A composition as defined in
claim 24
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
26. A composition as defined in
claim 23
, wherein said perturbant comprises a proteinoid.
27. A composition as defined in
claim 23
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
28. A composition as defined in
claim 46
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
29. A composition as defined in
claim 23
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
30. A composition as defined in
claim 23
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
31. A composition as defined in
claim 23
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2 to C10 alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
32. A dosage unit form comprising:
(A) a composition as defined in
claim 23
; and
(B)
(a) an excipient,
(b) a diluent,
(c) a disintegrant,
(d) a lubricant,
(e) a plasticizer,
(f) a colorant,
(g) a dosing vehicle, or
(h) any combination thereof.
33. A method for preparing an agent which is capable of being deliverable by the subcutaneous route to a subject in need of said agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a complexing perturbant to reversibly transform said biologically active agent to said intermediate state and to form a subcutaneously deliverable supramolecular complex,
said perturbant having a molecular weight between about 150 and about 600 daltons, and having at least one hydrophilic moiety and one hydrophilic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent,
said biologically active agent not forming a microsphere with said perturbant, and
said perturbant being present in an amount effective for subcutaneous delivery of said biologically active agent; and
(c) preparing a mimetic of said supramolecular complex.
34. A method as defined in
claim 33
, wherein said biologically active agent comprises a peptide and said mimetic comprises a peptide mimetic.
35. A method for preparing an agent which is capable of being delivered by the subcutaneous route to a subject in need of said agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a perturbant to reversibly transform said biologically active agent to said intermediate state, wherein said perturbant being present in an amount effective for subcutaneous delivery of said biologically active agent; and
(c) preparing a mimetic of said intermediate state.
36. A method as defined in
claim 35
, wherein said perturbant comprises a pH changing agent, an ionic strength changing agent, or guanidine hydrochloride.
37. A subcutaneous delivery composition comprising a mimetic of the subcutaneous delivery composition prepared by the method of
claim 13
.
38. A method for delivering, by the sublingual route, a biologically active agent to a subject in need of said biologically active agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a complexing perturbant to reversibility transform said biologically active agent to said intermediate state and to form a subcutaneously deliverable supramolecular complex,
said perturbant having a molecular weight between about 150 to about 600 daltons, and having at least one hydrophilic moiety and at least one hydrophobic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent,
said biologically active agent not forming a microsphere with said perturbant, and
said perturbant being present in an amount effective for sublingual delivery of said biologically active agent; and
(c) sublingually administering said supramolecular complex to said subject.
39. A method as defined in
claim 38
, further comprising
(d) after said administering step, removing said perturbant from said supramolecular complex to transform said biologically active agent to said native state.
40. A method as defined in
claim 39
, wherein step (d) comprises diluting said supramolecular complex.
41. A method as defined in
claim 38
, wherein said intermediate state has a ΔG ranging from about −20 kcal/mole to about 20 kcal/moles relative to said native state.
42. A method as defined in
claim 38
, wherein said biologically active agent is selected from the group consisting of a peptide, a mucopolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
43. A method as defined in
claim 42
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
44. A method as defined in
claim 38
, wherein said perturbant comprises a proteinoid.
45. A method as defined in
claim 38
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
46. A method as defined in
claim 38
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
47. A method as defined in
claim 38
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
48. A method as defined in
claim 38
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
49. A method as defined in
claim 38
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2 to C10alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
50. A method for preparing a sublingually deliverable biologically active agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states; and
(b) exposing said biologically active agent to a complexing perturbant to reversibility transform said biologically active agent to said intermediate state and to form a sublingually deliverable supramolecular complex,
said perturbant having a molecular weight ranging from about 150 to about 600 daltons, and having at least one hydrophilic moiety and at least one hydrophobic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent;
said biologically active agent not forming a microsphere with said perturbant; and
said perturbant being present in an amount effective for sublingual delivery of said biologically active agent.
51. A method as defined in
claim 50
, wherein said intermediate state has ΔG ranging from about −20 kcal/mole to about 20 kcal/moles relative to said native state.
52. A method as defined in
claim 50
, wherein said biologically active agent is selected from the group consisting of a peptide, a micropolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
53. A method as defined in
claim 52
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
54. A method as defined in
claim 50
, wherein said perturbant comprises a proteinoid.
55. A method as defined in
claim 50
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
56. A method as defined in
claim 50
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
57. A method as defined in
claim 50
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
58. A method as defined in
claim 50
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
59. A method as defined in
claim 50
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2 to C10alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
60. A sublingual delivery composition comprising a supramolecular complex comprising:
(a) a biologically active agent in an intermediate conformational state non-covalently complexed with
(b) a complexing perturbant having a molecular weight ranging from about 150 to about 600 and having at least one hydrophilic moiety and at least one hydrophobic moiety;
wherein said intermediate state is reversible to said native state and is conformationally between a native conformational and a denatured conformational state of said biologically active agent and said composition is not a microsphere; and said perturbant being present in an amount effective for sublingual delivery of said biologically active agent.
61. A composition as defined in
claim 60
, wherein said biologically active agent is selected from the group consisting of a peptide, a micropolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
62. A composition as defined in
claim 61
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
63. A composition as defined in
claim 60
, wherein said perturbant comprises a proteinoid.
64. A composition as defined in
claim 60
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
65. A composition as defined in
claim 60
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
66. A composition as defined in
claim 60
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
67. A composition as defined in
claim 60
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
68. A composition as defined in
claim 60
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2 to C10 alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
69. A dosage unit form comprising:
(A) a composition as defined in
claim 60
; and
(B)
(a) an excipient,
(b) a diluent,
(c) a disintegrant,
(d) a lubricant,
(e) a plasticizer,
(f) a colorant,
(g) a dosing vehicle, or
(h) any combination thereof.
70. A method for preparing an agent which is capable of being administered by the sublingual route to a subject in need of said agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a complexing perturbant to reversibly transform said biologically active agent to said intermediate state and to form a sublingually administrable supramolecular complex,
said perturbant having a molecular weight between about 150 and about 600 daltons, and having at least one hydrophilic moiety and one hydrophilic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent,
said biologically active agent not forming a microsphere with said perturbant; and
said perturbant being present in an amount effective for sublingual delivery of said biologically active agent; and
(c) preparing a mimetic of said supramolecular complex.
71. A method as defined in
claim 70
, wherein said biologically active agent comprises a peptide and said mimetic comprises a peptide mimetic.
72. A method for preparing an agent which is capable of being administered by the sublingual route to a subject in need of said agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a perturbant to reversibly transform said biologically active agent to said intermediate state, wherein said perturbant is in an amount effective for sublingual delivery of said biologically active agent; and
(c) preparing a mimetic of said intermediate state.
73. A method as defined in
claim 72
, wherein said perturbant comprises a pH changing agent, an ionic strength changing agent, or guanidine hydrochloride.
74. An oral delivery composition comprising a mimetic of the oral delivery composition prepared by the method of
claim 50
.
75. A method for delivering, by the intranasal route, a biologically active agent to a subject in need of said biologically active agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a complexing perturbant to reversibility transform said biologically active agent to said intermediate state and to form an intranasally administrable supramolecular complex,
said perturbant having a molecular weight between about 150 to about 600 daltons, and having at least one hydrophilic moiety and at least one hydrophobic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent,
said biologically active agent not forming a microsphere with said perturbant, and
said perturbant being present in an amount effective for intranasal delivery of said biologically active agent; and
(c) intranasally administering said supramolecular complex to said subject.
76. A method as defined in
claim 75
, further comprising
(d) after said administering step, removing said perturbant from said supramolecular complex to transform said biologically active agent to said native state.
77. A method as defined in
claim 76
, wherein step (d) comprises diluting said supramolecular complex.
78. A method as defined in
claim 75
, wherein said intermediate state has a ΔG ranging from about −20 kcal/mole to about 20 kcal/moles relative to said native state.
79. A method as defined in
claim 75
, wherein said biologically active agent is selected from the group consisting of a peptide, a mucopolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
80. A method as defined in
claim 79
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
81. A method as defined in
claim 75
, wherein said perturbant comprises a proteinoid.
82. A method as defined in
claim 75
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
83. A method as defined in
claim 75
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
84. A method as defined in
claim 75
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
85. A method as defined in
claim 75
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
86. A method as defined in
claim 75
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10alkyl)phenyl, (C2 to C10alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
87. A method for preparing an intranasally deliverable biologically active agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states; and
(b) exposing said biologically active agent to a complexing perturbant to reversibility transform said biologically active agent to said intermediate state and to form an intranasally administrable supramolecular complex,
said perturbant having a molecular weight ranging from about 150 to about 600 daltons, and having at least one hydrophilic moiety and at least one hydrophobic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent; and
said biologically active agent not forming a microsphere with said perturbant;
said perturbant being present in an amount effective for intranasal delivery of said biologically active agent.
88. A method as defined in
claim 87
, wherein said intermediate state has ΔG ranging from about −20 kcal/mole to about 20 kcal/moles relative to said native state.
89. A method as defined in
claim 87
, wherein said biologically active agent is selected from the group consisting of a peptide, a micropolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
90. A method as defined in
claim 89
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
91. A method as defined in
claim 87
, wherein said perturbant comprises a proteinoid.
92. A method as defined in
claim 87
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
93. A method as defined in
claim 87
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
94. A method as defined in
claim 87
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
95. A method as defined in
claim 87
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
96. A method as defined in
claim 87
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2 to C10 alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10 )alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
97. An intranasal delivery composition comprising a supramolecular complex comprising:
(a) a biologically active agent in an intermediate conformational state non-covalently complexed with
(b) a complexing perturbant having a molecular weight ranging from about 150 to about 600 and having at least one hydrophilic moiety and at least one hydrophobic moiety;
wherein said intermediate state is reversible to said native state and is conformationally between a native conformational and a denatured conformational state of said biologically active agent and said composition is not a microsphere; and said perturbant being present in an amount effective for intranasal delivery of said biologically active agent.
98. A composition as defined in
claim 97
, wherein said biologically active agent is selected from the group consisting of a peptide, a micropolysaccharide, a carbohydrate, a lipid, a pesticide, or any combination of the foregoing.
99. A composition as defined in
claim 98
, wherein said biologically-active agent is selected from the group consisting of human growth hormone, bovine growth hormone, growth hormone-releasing hormone, an interferon, interleukin-II, insulin, heparin, calcitonin, erythropoietin, atrial naturetic factor, an antigen, a monoclonal antibody, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, vasopressin, cromolyn sodium, vancomycin, desferrioxamine (DFO), or any combination of any of the foregoing.
100. A composition as defined in
claim 97
, wherein said perturbant comprises a proteinoid.
101. A composition as defined in
claim 97
, wherein said perturbant is selected from the group consisting of an acylated amino acid and an acylated poly amino acid.
102. A composition as defined in
claim 97
, wherein said perturbant is selected from the group consisting of a sulfonated amino acid and a sulfonated poly amino acid.
103. A composition as defined in
claim 97
, wherein said perturbant is selected from the group consisting of an acylated aldehyde of an amino acid and an acylated aldehyde of a poly amino acid.
104. A composition as defined in
claim 97
, wherein said perturbant is selected from the group consisting of an acylated ketone of an amino acid and an acylated ketone of a poly amino acid.
105. A composition as defined in
claim 97
, wherein said perturbant comprises a carboxylic acid having the formula
R—CO2H
wherein
R is C1 to C24 alkyl, C2 to C24 alkenyl, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, phenyl, naphthyl, (C1 to C10 alkyl)phenyl, (C2to C10 alkenyl)phenyl, (C1 to C10 alkyl)naphthyl, (C2 to C10 alkenyl)naphthyl, phenyl(C1 to C10 alkyl), phenyl(C2 to C10 alkenyl), naphthyl(C1 to C10 alkyl) and naphthyl(C2 to C10 alkenyl);
R being optionally substituted with C1 to C10 alkyl, C2 to C10 alkenyl, C1 to C4 alkoxy, —OH, —SH, —CO2R1, C3 to C10 cycloalkyl, C3 to C10 cycloalkenyl, heterocyclic having 3-10 ring atoms wherein the hetero atom is one or more atoms of N, O, S or any combination thereof, aryl, (C1 to C10 alkyl)aryl, aryl(C1 to C10)alkyl, or any combination thereof;
R being optionally interrupted by oxygen, nitrogen, sulfur, or any combination thereof; and
R1 is hydrogen, C1 to C4 alkyl or C2 to C4 alkenyl; or
a salt thereof.
106. A dosage unit form comprising:
(A) a composition as defined in
claim 97
; and
(B)
(a) an excipient,
(b) a diluent,
(c) a disintegrant,
(d) a lubricant,
(e) a plasticizer,
(f) a colorant,
(g) a dosing vehicle, or
(h) any combination thereof.
107. A method for preparing an agent which is capable of being administered by the intranasal route to a subject in need of said agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate conformational state which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a complexing perturbant to reversibly transform said biologically active agent to said intermediate state and to form an intranasally administrable supramolecular complex,
said perturbant having a molecular weight between about 150 and about 600 daltons, and having at least one hydrophilic moiety and one hydrophilic moiety,
said supramolecular complex comprising said perturbant non-covalently complexed with said biologically active agent,
said biologically active agent not forming a microsphere with said perturbant, and
said perturbant being present in an amount effective for intranasal delivery of said biologically active agent; and
(c) preparing a mimetic of said supramolecular complex.
108. A method as defined in
claim 107
, wherein said biologically active agent comprises a peptide and said mimetic comprises a peptide mimetic.
109. A method for preparing an agent which is capable of being administered by the intranasal route to a subject in need of said agent, said method comprising:
(a) providing a biologically active agent which can exist in a native conformational state, a denatured conformational state, and an intermediate which is reversible to said native state and is conformationally between said native and denatured states;
(b) exposing said biologically active agent to a perturbant to reversibly transform said biologically active agent to said intermediate state, wherein said perturbant is in an amount effective for intranasal delivery of said biologically active agent; and
(c) preparing a mimetic of said intermediate state.
110. A method as defined in
claim 109
, wherein said perturbant comprises a pH changing agent, an ionic strength changing agent, or guanidine hydrochloride.
111. An oral delivery composition comprising a mimetic of the oral delivery composition prepared by the method of
claim 87
.
US09/760,307 1990-08-02 2001-01-11 Active agent transport systems Abandoned US20010039258A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US09/760,307 US20010039258A1 (en) 1990-08-02 2001-01-11 Active agent transport systems

Applications Claiming Priority (17)

Application Number Priority Date Filing Date Title
SE9002558-6 1990-08-02
US89890992A 1992-06-15 1992-06-15
US07/920,346 US5443841A (en) 1992-06-15 1992-07-27 Proteinoid microspheres and methods for preparation and use thereof
US08/051,019 US5451410A (en) 1993-04-22 1993-04-22 Modified amino acids for encapsulating active agents
US08/076,803 US5578323A (en) 1992-06-15 1993-06-14 Proteinoid carriers and methods for preparation and use thereof
US14357193A 1993-10-26 1993-10-26
US08/168,776 US5447728A (en) 1992-06-15 1993-12-16 Desferrioxamine oral delivery system
PCT/US1994/004560 WO1994023767A1 (en) 1993-04-22 1994-04-22 Oral drug delivery compositions and methods
US08/231,622 US5629020A (en) 1994-04-22 1994-04-22 Modified amino acids for drug delivery
US08/231,623 US5541155A (en) 1994-04-22 1994-04-22 Acids and acid salts and their use in delivery systems
US08/315,200 US5693338A (en) 1994-09-29 1994-09-29 Diketopiperazine-based delivery systems
US08/316,404 US6331318B1 (en) 1994-09-30 1994-09-30 Carbon-substituted diketopiperazine delivery systems
US08/328,932 US5714167A (en) 1992-06-15 1994-10-25 Active agent transport systems
US08/763,183 US6099856A (en) 1992-06-15 1996-12-10 Active agent transport systems
US08/820,694 US6344213B1 (en) 1996-03-29 1997-03-18 Compounds and compositions for delivering active agents
US08/939,939 US6221367B1 (en) 1992-06-15 1997-09-29 Active agent transport systems
US09/760,307 US20010039258A1 (en) 1990-08-02 2001-01-11 Active agent transport systems

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/939,939 Continuation US6221367B1 (en) 1990-08-02 1997-09-29 Active agent transport systems

Publications (1)

Publication Number Publication Date
US20010039258A1 true US20010039258A1 (en) 2001-11-08

Family

ID=25473967

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/939,939 Expired - Fee Related US6221367B1 (en) 1990-08-02 1997-09-29 Active agent transport systems
US09/760,307 Abandoned US20010039258A1 (en) 1990-08-02 2001-01-11 Active agent transport systems

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/939,939 Expired - Fee Related US6221367B1 (en) 1990-08-02 1997-09-29 Active agent transport systems

Country Status (6)

Country Link
US (2) US6221367B1 (en)
EP (1) EP1021169A4 (en)
JP (1) JP2001517694A (en)
AU (1) AU735693B2 (en)
CA (1) CA2304951A1 (en)
WO (1) WO1999016427A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005112937A1 (en) 2004-05-19 2005-12-01 Emisphere Technologies, Inc. Acyclovir formulations
WO2006072070A2 (en) 2004-12-29 2006-07-06 Emisphere Technologies, Inc. Pharmaceutical formulations of gallium salts
US20070224262A1 (en) * 2004-05-06 2007-09-27 Shingai Majuru Solid Dosage Form of Wetted Heparin
US20070232537A1 (en) * 2005-12-19 2007-10-04 Nastech Pharmaceutical Company Inc. Intranasal pyy formulations with improved transmucosal pharmacokinetics
EP2279732A2 (en) 2004-05-14 2011-02-02 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
WO2011017346A2 (en) 2009-08-03 2011-02-10 Emisphere Technologies, Inc. Fast-acting naproxen composition with reduced gastrointestinal effects
US20130102530A1 (en) * 2000-12-29 2013-04-25 The Kenneth S. Warren Institute, Inc. Protection, Restoration and Enhancement of Erythropoietin-Responsive Cells, Tissues and Organs
US8771712B2 (en) 2006-05-09 2014-07-08 Emisphere Technologies, Inc. Topical administration of acyclovir
US8927015B2 (en) 2006-04-12 2015-01-06 Emisphere Technologies, Inc. Formulations for delivering insulin
US9364502B2 (en) 2006-06-28 2016-06-14 Emisphere Technologies, Inc. Gallium nitrate formulations

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6221367B1 (en) * 1992-06-15 2001-04-24 Emisphere Technologies, Inc. Active agent transport systems
US6916489B2 (en) * 1992-06-15 2005-07-12 Emisphere Technologies, Inc. Active agent transport systems
US6461643B2 (en) 1993-04-22 2002-10-08 Emisphere Technologies, Inc. Oral drug delivery compositions and methods
US6001347A (en) 1995-03-31 1999-12-14 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
AU2595697A (en) 1996-03-29 1997-10-22 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
DE69818049T2 (en) * 1997-06-23 2004-07-15 Tanabe Seiyaku Co., Ltd. INHIBITORS OF ALPHA4-BETA1 MEDIATED CELL ADHESION
US6440929B1 (en) 1998-07-27 2002-08-27 Emisphere Technologies, Inc. Pulmonary delivery of active agents
JP5144861B2 (en) 1998-07-27 2013-02-13 エミスフェアー・テクノロジーズ・インク Compounds and compositions for delivery of active agents
EP1102742B1 (en) * 1998-08-07 2006-06-14 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
CN1338924A (en) * 1999-01-08 2002-03-06 艾米斯菲尔技术有限公司 Polymeric delivery agents and delivery agent compounds
WO2000050386A1 (en) 1999-02-26 2000-08-31 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
NZ534409A (en) * 1999-04-05 2006-03-31 Emisphere Tech Inc Disodium salts, monohydrates, and ethanol solvates 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
US7279597B1 (en) 1999-11-05 2007-10-09 Emisphere Technologies, Inc. Phenyl amine carboxylic acid compounds and compositions for delivering active agents
US7151191B2 (en) * 2000-01-13 2006-12-19 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
KR100828668B1 (en) * 2000-06-29 2008-05-09 에미스페어 테크놀로지스, 인코포레이티드 Compounds and compositions for delivering active agents
US20030225300A1 (en) * 2001-04-19 2003-12-04 Emisphere Technologies Inc. Compounds and compositions for delivering active agents
WO2002089830A1 (en) * 2001-05-04 2002-11-14 North Carolina State University Polymer conjugates of insecticidal peptides or nucleic acids and methods of use thereof
US20030108585A1 (en) * 2001-05-04 2003-06-12 Roe R. Michael Polymer conjugates of insecticidal peptides or nucleic acids or insecticides and methods of use thereof
EP2409569B1 (en) * 2002-02-20 2017-08-16 Emisphere Technologies, Inc. Method for administering GLP-1 molecules
US7812120B2 (en) * 2003-03-21 2010-10-12 Par Pharmaceutical, Inc. Nasal calcitonin formulations containing chlorobutanol
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
NZ588373A (en) 2004-05-19 2012-01-12 Emisphere Tech Inc Topical cromolyn formulations
US8110547B2 (en) * 2005-01-12 2012-02-07 Emisphere Technologies, Inc. Compositions for buccal delivery of parathyroid hormone
WO2007011958A2 (en) 2005-07-15 2007-01-25 Emisphere Technologies, Inc. Intraoral dosage forms of glucagon
NZ566551A (en) * 2005-08-19 2011-09-30 Emisphere Tech Inc Cyclopropyl compounds and compositions for delivering active agents
JP5501758B2 (en) * 2007-03-16 2014-05-28 株式会社 資生堂 Anti-wrinkle / improving agent
AU2009239429B2 (en) 2008-04-21 2013-08-29 Otonomy, Inc. Auris formulations for treating otic diseases and conditions
US8784870B2 (en) 2008-07-21 2014-07-22 Otonomy, Inc. Controlled release compositions for modulating free-radical induced damage and methods of use thereof
EP2306975A4 (en) 2008-07-21 2012-10-31 Otonomy Inc Controlled-release otic structure modulating and innate immune system modulating compositions and methods for the treatment of otic disorders
KR101839864B1 (en) 2012-09-21 2018-03-20 인텐시티 쎄라퓨틱스, 인코포레이티드 Method of treating cancer
EP3328898B8 (en) 2015-07-28 2024-04-10 6452728 Canada Corp. Trkb or trkc agonist compositions and methods for the treatment of otic conditions
US10821185B2 (en) 2016-06-29 2020-11-03 Otonomy Inc. Triglyceride otic formulations and uses thereof

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4746675A (en) * 1983-04-04 1988-05-24 Teijin Limited External pharmaceutical composition
US4826817A (en) * 1986-02-07 1989-05-02 Brown Thomas E Amino acid and hydroxyamino acid transporter compounds for therapeutic applications, process and use
US4873087A (en) * 1982-01-14 1989-10-10 Toyo Jozo Company, Ltd. Suppository preparation having excellent absorption property
US5451410A (en) * 1993-04-22 1995-09-19 Emisphere Technologies, Inc. Modified amino acids for encapsulating active agents
US5629020A (en) * 1994-04-22 1997-05-13 Emisphere Technologies, Inc. Modified amino acids for drug delivery
US5714167A (en) * 1992-06-15 1998-02-03 Emisphere Technologies, Inc. Active agent transport systems
US6221367B1 (en) * 1992-06-15 2001-04-24 Emisphere Technologies, Inc. Active agent transport systems
US6242495B1 (en) * 1997-02-07 2001-06-05 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US20020001591A1 (en) * 1993-04-22 2002-01-03 Emisphere Technologies, Inc. Compositions for the delivery of antigens
US6344213B1 (en) * 1996-03-29 2002-02-05 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6346242B1 (en) * 1995-03-31 2002-02-12 Emishpere 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
US6384278B1 (en) * 2000-02-04 2002-05-07 Emisphere Technologies, Inc. Boron-mediated amidation of carboxylic acids
US6391303B1 (en) * 1996-11-18 2002-05-21 Emisphere Technologies, Inc. Methods and compositions for inducing oral tolerance in mammals
US6399798B2 (en) * 1999-02-05 2002-06-04 Emisphere Technologies, Inc. Method of preparing alkylated salicylamides
US6440929B1 (en) * 1998-07-27 2002-08-27 Emisphere Technologies, Inc. Pulmonary delivery of active agents
US20020120009A1 (en) * 1993-04-22 2002-08-29 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6623791B2 (en) * 1999-07-30 2003-09-23 Ppg Industries Ohio, Inc. Coating compositions having improved adhesion, coated substrates and methods related thereto
US6627228B1 (en) * 1999-01-08 2003-09-30 Emisphere Technologies, Inc. Polymeric delivery agents and delivery agents compounds
US6642411B1 (en) * 1998-07-27 2003-11-04 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6646162B2 (en) * 1999-02-26 2003-11-11 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6693208B2 (en) * 1999-12-16 2004-02-17 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6916489B2 (en) * 1992-06-15 2005-07-12 Emisphere Technologies, Inc. Active agent transport systems

Family Cites Families (167)

* 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
NL95043C (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
NL224379A (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
FR1351358A (en) 1958-12-22 1964-02-07 Ncr Co Process for forming impermeable coatings for particulate matter by liquid phase separation
FR1468601A (en) 1958-12-22 1967-02-10 Ncr Co Process for forming protective coatings for solid and liquid particles
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
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
IL36670A (en) 1971-04-21 1974-09-10 Sela M Therapeutic basic copolymers of amino acids
US3794561A (en) 1971-09-30 1974-02-26 Sasaki T Biologically active peptide and method of preparing the same
US3816404A (en) 1971-12-08 1974-06-11 Texaco Inc Preparation of caprolactam
US3933873A (en) 1971-12-08 1976-01-20 Texaco Inc. Preparation of omega-aminoalkanoic acids
US3795739A (en) 1972-02-14 1974-03-05 Hoffmann La Roche Treatment of parkinson disease
JPS5210427B2 (en) 1972-07-19 1977-03-24
CA1045977A (en) 1973-05-17 1979-01-09 Arthur D. Little Biodegradable, implantable drug delivery device, and process for preparing and using the same
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
DK140801B (en) 1975-01-15 1979-11-19 Nordisk Insulinlab Process for the preparation of a stable long-acting insulin preparation.
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
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
US4348384A (en) 1980-10-17 1982-09-07 Dainippon Pharmaceutical Co., Ltd. Pharmaceutical composition for oral administration containing coagulation factor VIII or IX
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
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
CA1262238C (en) 1982-09-30 1989-10-10 Human monoclonal antibodies against bacterial toxins
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
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
CS254355B1 (en) 1985-04-10 1988-01-15 Vladimir Saudek Soluble and biodegradatable copolymeres activated for bond of biologicaly active substances
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
EP0225130B1 (en) 1985-11-22 1991-10-30 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
KR950014440B1 (en) 1986-08-11 1995-11-28 이노바타 바이오메드 리미티드 Phamacentical formulation comprising microcapsules
US4837381A (en) 1986-08-11 1989-06-06 American Cyanamid Company Compositions for parenteral administration and their use
NL8720442A (en) 1986-08-18 1989-04-03 Clinical Technologies Ass DELIVERY SYSTEMS FOR PHARMACOLOGICAL AGENTS.
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
US5069936A (en) 1987-06-25 1991-12-03 Yen Richard C K Manufacturing protein microspheres
JPH0725725B2 (en) 1987-07-23 1995-03-22 保土谷化学工業株式会社 Benzamide derivative
MX12394A (en) 1987-07-23 1993-12-01 Ciba Geigy Ag PROCEDURE FOR OBTAINING POLYETHYLENE GLYCOL CARBAMATES.
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
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
GB8822857D0 (en) 1988-09-29 1988-11-02 Patralan Ltd Pharmaceutical formulations
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
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
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
FR2658076B1 (en) 1990-02-12 1992-06-12 Sanofi Sa COSMETIC COMPOSITION CONTAINING COPOLYMERS OF AMINO ACIDS, USEFUL AS A MOISTURIZING AGENT.
JPH05268986A (en) 1990-03-19 1993-10-19 Bristol Myers Squibb Co Monoclonal antibody and activation of lymphocyte
GB9007052D0 (en) 1990-03-29 1990-05-30 Skua Investments Ltd Pharmaceutical formulations
JP3249147B2 (en) 1990-06-01 2002-01-21 キリン−アムジエン・インコーポレーテツド Oral preparation containing bioactive protein
CA2046830C (en) 1990-07-19 1999-12-14 Patrick P. Deluca Drug delivery system involving inter-action between protein or polypeptide and hydrophobic biodegradable polymer
US5578323A (en) 1992-06-15 1996-11-26 Emisphere Technologies, Inc. Proteinoid carriers 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
US5443841A (en) 1992-06-15 1995-08-22 Emisphere Technologies, Inc. Proteinoid microspheres and methods for preparation and use thereof
US6099856A (en) * 1992-06-15 2000-08-08 Emisphere Technologies, Inc. Active agent transport systems
US5447728A (en) * 1992-06-15 1995-09-05 Emisphere Technologies, Inc. Desferrioxamine oral delivery system
US5693338A (en) 1994-09-29 1997-12-02 Emisphere Technologies, Inc. Diketopiperazine-based delivery systems
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
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
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
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
JPH08509464A (en) 1993-01-27 1996-10-08 セプラコア,インコーポレイテッド Methods and compositions using (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
US5709861A (en) 1993-04-22 1998-01-20 Emisphere Technologies, Inc. Compositions for the delivery of antigens
US5643957A (en) 1993-04-22 1997-07-01 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
DE69413590D1 (en) 1993-04-23 1998-11-05 Rhone Poulenc Chimie Polyanhydroaspartic acid and its biodegradable hydrolysis products
US5650386A (en) 1995-03-31 1997-07-22 Emisphere Technologies, Inc. Compositions for oral delivery of active agents
US5750147A (en) 1995-06-07 1998-05-12 Emisphere Technologies, Inc. Method of solubilizing and encapsulating itraconazole
US5667806A (en) 1995-06-07 1997-09-16 Emisphere Technologies, Inc. Spray drying method and apparatus

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4873087A (en) * 1982-01-14 1989-10-10 Toyo Jozo Company, Ltd. Suppository preparation having excellent absorption property
US4746675A (en) * 1983-04-04 1988-05-24 Teijin Limited External pharmaceutical composition
US4826817A (en) * 1986-02-07 1989-05-02 Brown Thomas E Amino acid and hydroxyamino acid transporter compounds for therapeutic applications, process and use
US6245359B1 (en) * 1992-06-15 2001-06-12 Emisphere Technologies, Inc. Active agent transport systems
US6348207B1 (en) * 1992-06-15 2002-02-19 Emisiphere Technologies, Inc. Orally deliverable supramolecular complex
US5714167A (en) * 1992-06-15 1998-02-03 Emisphere Technologies, Inc. Active agent transport systems
US6221367B1 (en) * 1992-06-15 2001-04-24 Emisphere Technologies, Inc. Active agent transport systems
US6916489B2 (en) * 1992-06-15 2005-07-12 Emisphere Technologies, Inc. Active agent transport systems
US20020001591A1 (en) * 1993-04-22 2002-01-03 Emisphere Technologies, Inc. Compositions for the delivery of antigens
US7005141B2 (en) * 1993-04-22 2006-02-28 Emisphere Technologies Inc. Oral drug delivery compositions and methods
US20020120009A1 (en) * 1993-04-22 2002-08-29 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6663887B2 (en) * 1993-04-22 2003-12-16 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US5451410A (en) * 1993-04-22 1995-09-19 Emisphere Technologies, Inc. Modified amino acids for encapsulating active agents
US6610329B2 (en) * 1993-04-22 2003-08-26 Emisphere Technologies Inc. Compositions for the delivery of antigens
US6461643B2 (en) * 1993-04-22 2002-10-08 Emisphere Technologies, Inc. Oral drug delivery compositions and methods
US5629020A (en) * 1994-04-22 1997-05-13 Emisphere Technologies, Inc. Modified amino acids for drug delivery
US6346242B1 (en) * 1995-03-31 2002-02-12 Emishpere Technologies, Inc. Compounds and compositions for delivering active agents
US6699467B2 (en) * 1995-03-31 2004-03-02 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6428780B2 (en) * 1995-03-31 2002-08-06 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6344213B1 (en) * 1996-03-29 2002-02-05 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6391303B1 (en) * 1996-11-18 2002-05-21 Emisphere Technologies, Inc. Methods and compositions for inducing oral tolerance in mammals
US6358504B1 (en) * 1997-02-07 2002-03-19 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6525020B2 (en) * 1997-02-07 2003-02-25 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6242495B1 (en) * 1997-02-07 2001-06-05 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
US6440929B1 (en) * 1998-07-27 2002-08-27 Emisphere Technologies, Inc. Pulmonary delivery of active agents
US6642411B1 (en) * 1998-07-27 2003-11-04 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6693073B2 (en) * 1998-07-27 2004-02-17 Emisphere Technologies, Inc. Pulmonary delivery of active agents
US6627228B1 (en) * 1999-01-08 2003-09-30 Emisphere Technologies, Inc. Polymeric delivery agents and delivery agents compounds
US6399798B2 (en) * 1999-02-05 2002-06-04 Emisphere Technologies, Inc. Method of preparing alkylated salicylamides
US6646162B2 (en) * 1999-02-26 2003-11-11 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6623791B2 (en) * 1999-07-30 2003-09-23 Ppg Industries Ohio, Inc. Coating compositions having improved adhesion, coated substrates and methods related thereto
US6693208B2 (en) * 1999-12-16 2004-02-17 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US6384278B1 (en) * 2000-02-04 2002-05-07 Emisphere Technologies, Inc. Boron-mediated amidation of carboxylic acids

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130102530A1 (en) * 2000-12-29 2013-04-25 The Kenneth S. Warren Institute, Inc. Protection, Restoration and Enhancement of Erythropoietin-Responsive Cells, Tissues and Organs
US20070224262A1 (en) * 2004-05-06 2007-09-27 Shingai Majuru Solid Dosage Form of Wetted Heparin
US8039018B2 (en) 2004-05-06 2011-10-18 Emisphere Technologies, Inc. Solid dosage form of wetted heparin
EP2279732A2 (en) 2004-05-14 2011-02-02 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
WO2005112937A1 (en) 2004-05-19 2005-12-01 Emisphere Technologies, Inc. Acyclovir formulations
WO2006072070A2 (en) 2004-12-29 2006-07-06 Emisphere Technologies, Inc. Pharmaceutical formulations of gallium salts
US20070232537A1 (en) * 2005-12-19 2007-10-04 Nastech Pharmaceutical Company Inc. Intranasal pyy formulations with improved transmucosal pharmacokinetics
US8927015B2 (en) 2006-04-12 2015-01-06 Emisphere Technologies, Inc. Formulations for delivering insulin
US8771712B2 (en) 2006-05-09 2014-07-08 Emisphere Technologies, Inc. Topical administration of acyclovir
US9364502B2 (en) 2006-06-28 2016-06-14 Emisphere Technologies, Inc. Gallium nitrate formulations
WO2011017346A2 (en) 2009-08-03 2011-02-10 Emisphere Technologies, Inc. Fast-acting naproxen composition with reduced gastrointestinal effects
US20110039930A1 (en) * 2009-08-03 2011-02-17 Emisphere Technologies, Inc. Fast-acting naproxen composition with reduced gastrointestinal effects

Also Published As

Publication number Publication date
EP1021169A4 (en) 2004-07-07
EP1021169A1 (en) 2000-07-26
AU735693B2 (en) 2001-07-12
WO1999016427A1 (en) 1999-04-08
US6221367B1 (en) 2001-04-24
AU9513698A (en) 1999-04-23
CA2304951A1 (en) 1999-04-08
JP2001517694A (en) 2001-10-09

Similar Documents

Publication Publication Date Title
US6221367B1 (en) Active agent transport systems
US6245359B1 (en) Active agent transport systems
US6099856A (en) Active agent transport systems
US20050255138A1 (en) Active agent transport systems
US5863944A (en) Compounds and compositions for delivering active agents
US7005141B2 (en) Oral drug delivery compositions and methods
ES2244367T3 (en) COMPOSITIONS FOR ADMINISTRATION VIA ORAL.
JP4975201B2 (en) Compositions for delivering compounds and active agents
JP2003313157A (en) Compound and composition for delivering active agent
CA2202300C (en) Active agent transport systems
MXPA00003109A (en) Active agent transport systems
JP5215977B2 (en) Compositions for delivering compounds and active agents
MXPA99009632A (en) Compounds and compositions for delivering active agents

Legal Events

Date Code Title Description
AS Assignment

Owner name: MHR INSTITUTIONAL PARTNERS IIA LP,NEW YORK

Free format text: SECURITY AGREEMENT;ASSIGNOR:EMISPHERE TECHNOLOGIES, INC.;REEL/FRAME:016617/0145

Effective date: 20050926

Owner name: MHR INSTITUTIONAL PARTNERS IIA LP, NEW YORK

Free format text: SECURITY AGREEMENT;ASSIGNOR:EMISPHERE TECHNOLOGIES, INC.;REEL/FRAME:016617/0145

Effective date: 20050926

STCB Information on status: application discontinuation

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