US20090280169A1 - Compositions of peptides and processes of preparation thereof - Google Patents

Compositions of peptides and processes of preparation thereof Download PDF

Info

Publication number
US20090280169A1
US20090280169A1 US12/436,990 US43699009A US2009280169A1 US 20090280169 A1 US20090280169 A1 US 20090280169A1 US 43699009 A US43699009 A US 43699009A US 2009280169 A1 US2009280169 A1 US 2009280169A1
Authority
US
United States
Prior art keywords
composition
derivative
peptide
polypeptide
protein
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
US12/436,990
Inventor
Thomas W. Leonard
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.)
Novo Nordisk AS
Original Assignee
Merrion Research Ill Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merrion Research Ill Ltd filed Critical Merrion Research Ill Ltd
Priority to US12/436,990 priority Critical patent/US20090280169A1/en
Priority to TW098115182A priority patent/TW200950801A/en
Assigned to MERRION RESEARCH III LIMITED reassignment MERRION RESEARCH III LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEONARD, THOMAS W.
Publication of US20090280169A1 publication Critical patent/US20090280169A1/en
Assigned to NOVO NORDISK A/S reassignment NOVO NORDISK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MERRION RESEARCH III LIMITED
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • A61K38/09Luteinising hormone-releasing hormone [LHRH], i.e. Gonadotropin-releasing hormone [GnRH]; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/02Peptides of undefined number of amino acids; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/03Peptides having up to 20 amino acids in an undefined or only partially defined sequence; Derivatives thereof
    • 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
    • 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
    • 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
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • A61K9/2846Poly(meth)acrylates
    • 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
    • A61K9/4891Coated capsules; Multilayered drug free capsule shells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/16Masculine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/18Feminine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/02Drugs for disorders of the endocrine system of the hypothalamic hormones, e.g. TRH, GnRH, CRH, GRH, somatostatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids

Definitions

  • the present invention generally relates to compositions of peptides, polypeptides, proteins, analogues or derivatives thereof, and the process of preparation thereof.
  • a variety of biologically active peptides, polypeptides and proteins have been widely used for medical treatment. For a majority of medical treatments, there is a need to deliver a sustained level of biologically active peptides or proteins to animals or humans to provide a stable therapeutic effect. Additionally, the biologically active peptides or proteins also need to be stored for a prolonged period of time and still maintain their activity. However, many naturally occurring and synthetic peptides and proteins as well as their analogs have exhibited a tendency to form gels, aggregates, fibrils, dimers, other polymers, coagulates, etc. In some cases, the formed material may be able to revert back to the active monomer form. In other cases, the altered state may be permanent and represent a degraded state.
  • Some exemplary biologically active peptides include insulin, glucagon-like peptide-1 (GLP-1) and Gonadotropin-releasing hormone (GnRH) analogs.
  • GLP-1 glucagon-like peptide-1
  • GnRH Gonadotropin-releasing hormone
  • U.S. Pat. No. 6,124,261 describes a non aqueous polar aprotic peptide formulation that may be used to stabilize the peptide.
  • a citrate buffering agent may be used to reduce the gelation of fatty acid-acylated protein. See U.S. Pat. No. 5,631,347.
  • compositions comprising a therapeutically effective amount of at least one peptide, polypeptide, protein, an analog or derivative thereof and a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof in a solution, wherein at least one stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent.
  • at least one peptide, polypeptide, protein, an analog or derivative thereof is insulin, or an analog or derivative thereof.
  • At least one peptide, polypeptide, protein, analog or derivative thereof is Glucagon-Like Peptide (GLP), or an analog or derivatives thereof.
  • the stabilizing agent is selected from the group consisting of sodium caprylate, sodium caprate and sodium laurate.
  • the methods comprises mixing the peptide, polypeptide, protein, analog or derivative thereof with a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof, and the stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms.
  • the stabilizing agent is a medium chain fatty acid salt has a carbon chain length of from about 8 to about 14 carbon atoms.
  • FIG. 1 is a table describing the preparation of different acyline batches.
  • FIG. 2 shows appearance of 9% and 16.7% acyline samples of different acyline batches in propylene glycol.
  • FIG. 3 shows the appearance of 0.5% and 1% acyline samples of different acyline batches in water.
  • FIG. 4( a ) shows the appearance of 5 mg and 10 mg doses acyline microemulsions of different acyline batches.
  • FIG. 4( b ) shows the formulation of the microemulsion.
  • FIG. 5 graphically demonstrates the comparison results of the gelation of 0.1 mg/mL acyline sample with 1% and 0% Tween 80.
  • FIG. 6 graphically demonstrates the comparison results of the gelation of 0.1 mg/mL acyline sample having 5 mg/mL, 1 mg/mL, 0.1 mg/mL and 1% Tween 80.
  • FIG. 7 graphically demonstrates the comparison results of the gelation of 0.1 and 0.01 mg/mL acyline sample having 1% and 1 mg/mL Tween 80.
  • FIG. 8 graphically demonstrates the comparison results of the gelation of 0.01 mg/mL acyline sample having 0.1%, 0.5% and 1% Tween 80.
  • FIG. 9 graphically demonstrates the correlation between the concentration of sodium caprate and the gelation of acyline.
  • FIG. 10( a ) illustrates the formulation of microemulsion 1 —55% Capmul MCM.
  • FIG. 10( b ) illustrates the formulation of microemulsion 2 —45% Capmul PG-8.
  • FIG. 10( c ) illustrates the formulation of microemulsion 3 —55% Capmul MCM C10.
  • FIG. 11 graphically demonstrates the comparison results of relative bioavailability of different formulations of acyline.
  • FIG. 12 graphically demonstrates the comparison results of relative bioavailability of (1) enteric tablets 10 mg acyline versus acyline sample with no surface active agent and (2) enteric tablets 10 mg acyline versus 5 mg acyline with sodium caprate sample.
  • FIG. 13( a ) illustrates the correlation of % of recovery of insulin for insulin formulations with or without sodium caprate.
  • FIG. 13( b ) illustrates % of recovery of insulin from insulin formulations with capric acid.
  • alcohol means an organic compound in which one or more hydroxyl (OH) groups are attached to carbon (C) atoms in place of hydrogen (H) atoms.
  • the alcohol contains 1-6 carbon atoms.
  • the alcohol contains 1-4 carbon atoms.
  • Exemplary alcohols include, but are not limited to, methanol, ethanol, n-propanol, iso-propanol, butanol, tert-butanol, pentanol, hexanol.
  • gelation means that a compound of interest undergoes aggregation to form fibrils, dimers, longer polymers, coagulates, or structures that may result in formation of a colloid structure or gel.
  • the viscosity of the mixture, used in the present application may be applied to a compound of interest in aqueous solution or a solid mass.
  • “Gelation” may occur to varying extents, and may occur in such a maimer as to be non-detectable by ordinary means. For example, there may be no increased viscosity or changes in the flow characteristics of the solution.
  • the material formed from the gelation (hereinafter “gel”) may be removed by physical means such as filtration and thus detected by analytical techniques known to one skilled in the art. The presence of gels may cause significant problems in the development of different administration forms. For example, when the gelled system is processed to obtain powders of the drug as part of drug substance manufacturing techniques, a xerogel may be formed during the process.
  • ‘xerogel’ is a solid formed from the gel after the liquid is removed from a gelled system.
  • the powder obtained from the gelled system containing xerogel may have substantially different characteristics from powders obtained from solutions that contain no gelled material. In some situations, obtained power loses the biological activity. Therefore, the formation of gels may potentially cause deleterious effects at either the drug substance processing stage, or at the stage of preparing the dosage form or storage stage.
  • the degree of agglomeration or coalescence which results in the formation of the gel may be measured by various methods known to one skilled in the art, for example micro or macro filtration followed by assay of the filtrate, centrifugation followed by assay of the supernatant, or various optical absorption or diffraction methods including those utilizing visible or UV light methods, or laser based methods.
  • the methods of measuring physical characteristics of the liquid may also be used such as surface tension, freezing point depression, viscosity, or measurement of other colligative properties.
  • the degree of xerogel formation may be measured by dispersing the material in water and using the techniques described herein or by direct measurement on the powder by methods known to one skilled in the art including x-ray powder diffraction, IR spectroscopy, or other methods known to the art that may be carried out on powdered materials.
  • the term “reduce the gelation” means to disrupt, retard or eliminate the gel formation of a compound. In situation where the gelation is reversible, the term “reduce the gelation” also means to reverse the gel back to monomeric form of the compound, which maintains it biological activity.
  • the term “anti-gelling agent” refers to an agent, a compound, a composition or a combination thereof which may inhibit the gelation of at least 50% of the compound of interest, when a sufficient amount of the anti-gelling agent is used. In some embodiments, the anti-gelling agent may inhibit the gelation of at least 80% of the compound of interest, when a sufficient amount of the anti-gelling agent is used. In some embodiments, the anti-gelling may inhibit the gelation of at least 90% of the compound of interest, when a sufficient amount of the anti-gelling agent is used.
  • a “therapeutically effective” or “therapeutically acceptable” amount refers to an amount that will elicit a therapeutically useful response in a subject.
  • the therapeutically useful response may provide some alleviation, mitigation, or decrease in at least one clinical symptom in the subject.
  • the therapeutic useful response need not be complete or curative, as long as some benefit is provided to the subject.
  • the subject is an animal. In some embodiments, the subject is a human.
  • water miscible solvent means a solvent that can be mixed with water to form a solution. Water miscible solvents may be used to create a hydrophilic phase.
  • compositions comprising, consisting essentially of, or consisting of, a therapeutically effective amount of at least one peptide, polypeptide, protein, an analog or derivative thereof and a sufficient amount of at least one stabilizing agents to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof.
  • a “stabilizing agent” is an agent that improves the stability of a peptide, polypeptide, protein, an analog or derivative thereof described herein.
  • At least one stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms. In some embodiments, at least one stabilizing agent is a surface active agent.
  • the present invention provides compositions that maintain the stability of peptide, polypeptide, protein, an analog or derivative thereof described herein for a sufficient time to be stored, processed and/or administered for treatment.
  • One way of measuring the stability is measuring the percentage of peptide, polypeptide, protein, an analog or derivative thereof is retained under a specific condition.
  • the term “peptide, polypeptide, protein, analog or derivative thereof is retained” means that the peptide, polypeptide, protein, an analogs or derivative thereof does not form gels, aggregates, fibrils, dimers, other polymers, coagulates, rather it maintains as a monomer or its biological activity after a period of time.
  • composition of the present invention at least about 50% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, in the composition of the present invention, at least about 80% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, in the composition of the present invention, at least about 90% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours.
  • composition of the present invention at least about 95% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours.
  • the stability is improved when the gelation of peptide, polypeptide, protein, an analog or derivative thereof is reduced.
  • compositions described herein may be used directly for storage, processing or administration.
  • the composition may also be used to mix with a suitable medium (e.g. a solvent, a microemulsion or a solid mass) for storage, processing or administration.
  • a suitable medium e.g. a solvent, a microemulsion or a solid mass
  • the stability of peptide, polypeptide, protein, analogues or derivatives thereof in the medium is improved as described herein.
  • the stabilizing agent is an anti-gelling agent.
  • the present invention provides compositions comprising, a therapeutically effective amount of one or more peptide, polypeptide, protein, an analog or derivative thereof, and a sufficient amount of at least one anti-gelling agents to reduce the gelation of the peptide, polypeptide, protein, an analog or derivative thereof.
  • the dissolution rate of the stabilizing agent and the peptide, polypeptide, protein, analogs or derivatives thereof in the composition are substantially the same.
  • the present invention can be applied to any peptide, polypeptide, protein, analog or derivative that has a tendency to aggregates, dimerizes, polymerizes, coagulates, gels or fibrillates. Analogs, derivatives, and pharmaceutically acceptable salts of any of the peptides, polypeptides or proteins are included in these terms.
  • the “tendency to to aggregates, dimerize, polymerize, coagulate, gel or fibrillate” refers to that at least 50% of a compound of interest undergoes aggregation to form fibrils, dimers, polymers that coagulates, or structures that may result in formation of a colloid structure or gel in a system at a certain temperature (e.g. 37° C.) after the system stands for a period of time (e.g. at least about 24 hours).
  • At least one peptide, polypeptide, protein, an analog or derivative thereof is an insulin or an analog or derivative thereof.
  • At least one peptide, polypeptide, protein, analogs or derivatives thereof is Glucagon-Like Peptide (GLP), or an analog or derivative thereof.
  • GLP Glucagon-Like Peptide
  • at least one GLP is selected from the group consisting of GLP-1, GLP-1 analogs, derivatives thereof, GLP-2, GLP-2 analogs, derivatives thereof, and exendin-4, analogs and derivatives thereof.
  • More exemplary peptides, polypeptides, proteins, analogs or derivatives thereof include, but are not limited to, GnRH agonists and antagonists, somatostatin, ACTH, corticotropin-releasing factor, angiotensin, calcitonin, gastric inhibitory peptide, growth hormone, growth hormone-releasing factor, pituitary adenylate, exendin, exendin-3, cyclase activating peptide, secretin, enterogastrin, somatostatin, somatotropin, somatomedin, parathyroid hormone, thrombopoietin, erythropoietin, hypothalamic releasing factors, prolactin, thyroid stimulating hormones, endorphins, enkephalins, vasopressin, oxytocin, opioids and analogues thereof, superoxide dismutase, interferon, asparaginase, arginase, arginine deaminase,
  • At least one peptide, polypeptide, protein, analog or derivative comprises a variety of GnRH related compounds which have a tendency of gelation.
  • the “tendency of gelation” refers to that at least 50% of a compound undergoes aggregation to form fibrils, dimers, polymers that coagulates, or structures that may result in formation of a colloid structure or gel in a system at a certain temperature (e.g. 37° C.) after the system stands for a period of time (e.g. at least about 2 hours).
  • GnRH related compounds include both GnRH antagonists and GnRH agonists. In some embodiments, the present invention may be applied to GnRH antagonists.
  • the present invention include, but is not limited to, the following GnRH antagonists, acyline (Ac-D2Nal-D4Cpa-D3Pal-Ser4Aph(Ac)-D4Aph(Ac)-Leu-ILys-Pro-DAla-NH 2 ), Acetyl- ⁇ -[2-Naphthyl]-D-Ala-D-p-Chloro-Phe- ⁇ -[3-Pyridyl]-D-Ala-Ser-N ⁇ -[Nicotinoyl]-Lys-N ⁇ -[Nicotinoyl]-D-Lys-Leu-N ⁇ -[Isopropyl]-Lys-Pro-D-Ala-NH 2 (also referred to herein as Antide), acetyl-D2Nal1, D4CIPhe2, D3Pal3, ARg5, Dglu6 (AA) (also referred to herein as Nal
  • the GnRH antagonists of the present invention have a tendency of gelation in the presence of ions.
  • At least one GnRH antagonist is selected from the group consisting of acyline, abarelix, azaline B, cetrorelix, ganirelix, teverelix, degarelix, antide, orntide and GnRH antagonists described in U.S. Pat. No. 7,098,305.
  • At least one GnRH antagonist is selected from the group consisting of abarelix, cetrorelix, degarelix, ganirelix, and a pharmaceutically acceptable salt thereof.
  • abarelix refers to a compound having a structure of Formula I
  • the IUPAC name of Formula I is acetyl-D- ⁇ -naphthylalanyl-D-4-chlorophenylalanyl-D-3-pyridylalanyl-L-seryl-L-N-methyl-tyrosyl-D-asparagyl-L-leucyl-L-N(e)-isopropyl-lysyl-L-prolyl-D-alanyl-amide.
  • abarelix includes the compound of Formula I, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these.
  • abarelix also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula I and pharmaceutically acceptable salts thereof.
  • cetrorelix refers to a compound having a structure of Formula II.
  • the IUPAC name of Formula II is Acetyl-D-3-(2′-naphtyl)-alanine-D-4-chlorophenylalanine-D-3-(3′-pyridyl)-alanine-L-serine-L-tyrosine-D-citruline-L-leucine-L-arginine-L-proline-D-alanine-amide.
  • cetrorelix includes the compound of Formula II, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these.
  • cetrorelix also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula II and pharmaceutically acceptable salts thereof.
  • the term “degarelix” refers to a compound having a structure of Formula III.
  • the IUPAC name of Formula III is N-acetyl-3-(naphtalen-2-yl)-D-alanyl-4-chloro-D-phenylalanyl-3-(pyridin-3-yl)-D-alanyl-L-seryl-4-(((4S)-2,6-dioxohexahydropyrimidin-4-yl)carbonyl)amino)-L-phenylalanyl-4-(carbamoylamino)-D-phenylalanyl-L-leucyl-N6-(1-methylethyl)-L-lysyl-L-prolyl-D-alaninamide.
  • degarelix includes the compound of Formula III, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these.
  • degarelix also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula III and pharmaceutically acceptable salts thereof.
  • ganirelix refers to a compound having a structure of Formula IV.
  • the IUPAC name of Formula IV is a (2S)-1-[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2R)-2-[[(2S)-2-[[(2R)-2-[[(2R)-2-acetamido-3-naphthalen-2-ylpropanoyl]amino]-3-(4-chlorophenyl)propanoyl]amino]-3-pyridin-3-ylpropanoyl]amino]-3-hydroxypropanoyl]amino]-3-(4-hydroxyphenyl)propanoyl]amino]-6-[bis(ethylamino)methylideneamino]hexanoyl]amino]-4-methylpentanoyl]amino]-6-[bis(ethylamino)methylideneamino]hexanoyl]-N-[(
  • ganirelix includes the compound of Formula IV, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these.
  • ganirelix also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula IV and pharmaceutically acceptable salts thereof.
  • the present invention may be applied to GnRH agonists that have a tendency of gelation.
  • the exemplary GnRH agonists include, but are not limited to, histerelin, leuprolide and goserelin.
  • GnRH related compound include all forms thereof including stereoisomers, enantiomers, diastereomers, racemic mixtures, and derivatives thereof, for example, salts, acids, esters and the like.
  • the compound may be provided in any suitable phase state including as solid, liquid, solution, suspension and the like. When provided in a solid particulate form, the particles may be of any suitable size or morphology and may assume one or more crystalline, semi-crystalline and/or amorphous forms.
  • the peptide, polypeptide, protein, analogs or derivatives there of used in the present invention may be present in any amount which is sufficient to elicit a therapeutic effect and, where applicable, may be present either substantially in the form of one optically pure enantiomer or as a mixture, racemic or otherwise, of enantiomers.
  • the actual amount of peptide, polypeptide, protein, analogs or derivatives there of used in the composition will depend on the potency of the selected compound in question.
  • peptides, polypeptides, proteins, analogs or derivatives described herein may be obtained through commercial resources or may be prepared according to methods known to one skill in the art.
  • the GnRH antagonists applied in the present invention may be prepared using a method known to one of ordinary skill in the art or a process described in the present invention.
  • acyline can be prepared according to the method described in U.S. Pat. No. 5,506,207.
  • the amount of the stabilizing agent used in the present invention can vary considerably. For example, the amount can be dependent upon individual stabilizing agents, solvent systems and other components in the composition. Generally, the amount of stabilizing agent should be sufficient to improve the stability of peptide, polypeptide, protein, analogs or derivatives thereof in a system such that at least 50% of peptide, polypeptide, protein, an analog or derivative thereof is retained when the composition stands at 37° C. for at least about 24 hours. In some embodiments, the amount of stabilizing agent should be sufficient to improve the stability of peptide, polypeptide, protein, analogs or derivatives thereof in a system such that at least 80% of peptide, polypeptide, protein, analog or derivative thereof is retained when the composition stands at 37° C.
  • the amount of anti-gelling agent should be sufficient to be sufficient to improve the stability of peptide, polypeptide, protein, an analog or derivative thereof in a system such that at least 90% of peptide, polypeptide, protein, an analog or derivative thereof is retained when the composition stands at 37° C. for at least about 24 hours.
  • the concentration of the stabilizing agent in the composition is at least equal to or above the CMC (Critical Micelle Concentration) of the stabilizing agent in the composition.
  • At least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 4 to 20 carbon atoms. In some embodiments, at least one stabilizing agent is medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms. In some embodiments, the carbon chain length is from 8 to 14. In some embodiments, at least one stabilizing agent is a salt of medium chain fatty acid and has a carbon chain length of from 8 to 14 carbon atoms.
  • At least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms; with the provisos that (i) where the stabilizing agent is an ester of a medium chain fatty acid, said chain length of from 6 to 20 carbon atoms relates to the chain length of the carboxylate moiety, and (ii) where the stabilizing agent is an ether of a medium chain fatty acid, at least one alkoxy group has a carbon chain length of from 6 to 20 carbon atoms.
  • At least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid which is solid at room temperature and which has a carbon chain length of from 8 to 14 carbon atoms; with the provisos that (i) where the stabilizing agent is an ester of a medium chain fatty acid, said chain length of from 8 to 14 carbon atoms relates to the chain length of the carboxylate moiety, and (ii) where the stabilizing agent is an ether of a medium chain fatty acid, at least one alkoxy group has a carbon chain length of from 8 to 14 carbon atoms.
  • At least one stabilizing agent is a sodium salt of a medium chain fatty acid, the medium chain fatty acid having a carbon chain length of from 8 to 14 carbon atoms.
  • the stabilizing agent is solid at room temperature.
  • at least one stabilizing agent is selected from the group consisting of sodium caprylate, sodium caprate, and sodium laurate. In one embodiment, at least one stabilizing agent is sodium caprate.
  • At least one stabilizing agent is a surface active agent.
  • surface active agent refers to an agent that lowers the surface tension of the medium in which it is dissolved and/or the interfacial tension with other phases, and, accordingly, is positively adsorbed at the liquid/vapor and/or at other interfaces.
  • the surface active agents employed in the present invention include both ionic agents, i.e., cationic, anionic or zwitterionic, and non-ionic agents, or a mixture thereof.
  • cationic surface active agents include, but are not limited to, benzalkonium chloride, dicetyl ammonium chloride, cetyldimethylethylammonium bromide, cetylpyridinium chloride and salts of the above surface active agents.
  • anionic surface active agents include, but are not limited to, sodium stearoyl lactylate, hydrogenated lecithin, sodium lauryl sulfate, C 8-32 fatty acids and salts thereof, cholic acid and derivatives thereof such as deoxycholate, and its salts, ursodeoxycholic acid, and taurocholic acid; C 8-56 diesters of tartaric acid; phospholipids such as phosphatidic acid and phosphatidyl serine; C 5-29 monoesters of lactic acid; C 8-20 sulfonates, including alkyl-, olefin-, and alkylaryl derivatives; tridecyl- and dodecylbenzene sulfonic acids; and C 5-33 sarcosine and betaine derivatives.
  • zwitterionic surface active agents include, but are not limited to, phospholipids such as lecithin, phosphatidylethanolamine, sphingomyelins, dodecyl betaine, dodecyl dimethylamine oxide, cocamidopropyl betaine, and coco ampho glycinate.
  • non-ionic surface active agents include, but are not limited to, steareths; polyethylene glycol (PEGs); polysorbates (e.g. Tween 80); cetearyl glucoside; various commercially available sorbitans and their derivatives, for example, sorbitan hexastearate ethoxylate EO 6 mole, sorbitan isostearate, sorbitan laurate, sorbitan monoisostearate ethoxylate EO 20 mole, sorbitan monolaurate ethoxylate EO 20 mole, sorbitan monooleate ethoxylate EO 20 mole, sorbitan monopalmitate ethoxylate EO 20 mole, sorbitan monostearate ethoxylate EO 20 mole, sorbitan monstearate ethoxylate EO 6 mole, Sorbitan oleate, sorbitan palmitate, sorbitan sesquioleate, sorbitan ste
  • At least one surface active agent is selected from the group consisting of sodium lauryl sulfate, polysorbate surface active agents (such as polysorbate 20 (Tween 20), polysorbate 80 (Tween 80)), sorbitan surface active agents, sorbitan monolaurate and polyethoxylated castor oil and a combination thereof.
  • at least one surface active agent comprises polysorbate.
  • the composition of the present invention further comprises one or more excipients.
  • excipients are selected from the group consisting of rate-controlling polymeric materials, diluents, lubricants, disintegrants, plasticizers, anti-tack agents, opacifying agents, pigments, and flavorings.
  • rate controlling polymer material comprises hydrophilic polymers, hydrophobic polymers and mixtures of hydrophilic and/or hydrophobic polymers that are capable of controlling or retarding the release of the drug compound such as a GnRH related compound from a solid oral dosage form of the present invention.
  • Suitable rate controlling polymer materials include those selected from the group consisting of hydroxyalkyl cellulose such as hydroxypropyl cellulose and hydroxypropyl methyl cellulose; poly(ethylene) oxide; alkyl cellulose such as ethyl cellulose and methyl cellulose; carboxymethyl cellulose, hydrophilic cellulose derivatives; polyethylene glycol; polyvinylpyrrolidone; cellulose acetate; cellulose acetate butyrate; cellulose acetate phthalate; cellulose acetate trimellitate; polyvinyl acetate phthalate; hydroxypropylmethyl cellulose phthalate; hydroxypropylmethyl cellulose acetate succinate; polyvinyl acetaldiethylamino acetate; poly(alkylmethacrylate) and poly (vinyl acetate).
  • Suitable hydrophobic polymers include polymers and/or copolymers derived from acrylic or methacrylic acid and their respective esters, zein, waxes, shellac and hydrogenated vegetable oils.
  • the rate-controlling polymer comprises a polymer derived from acrylic or methacrylic acid and their respective esters or copolymers derived from acrylic or methacrylic acid and their respective esters.
  • the rate-controlling polymer comprises hydroxypropylmethylcellulose (HPMC).
  • At least one rate controlling polymer material is selected from the group consisting of poly acrylic acid, poly acrylate, poly methacrylic acid and poly methacrylate polymers such as those sold under the Eudragit® trade name (Rohm GmbH, Darmstadt, Germany), for example, Eudragit® L, Eudragit® S, Eudragit® RL, Eudragit® RS coating materials and mixtures thereof. Some of these polymers can be used as delayed release polymers to control the site where the drug is released.
  • the rate controlling polymer include polymethacrylate polymers such as those sold under the Eudragit® trade name (Rohm GmbH, Darmstadt, Germany), for example, Eudragit® L, Eudragit® S, Eudragit® RL, Eudragit® RS coating materials and mixtures thereof.
  • the present invention may further comprise diluents.
  • diluents Any suitable diluent may be used in the present invention.
  • exemplary diluents include, but are not limited to, pharmaceutically acceptable inert fillers such as microcrystalline cellulose, lactose, dibasic calcium phosphate, saccharides, and/or mixtures thereof.
  • diluents include microcrystalline cellulose such as those sold under the Avicel trademark (FMC Corp., Philadelphia, Pa.), for example, AvicelTM pH101, AvicelTM pH102 and AvicelTM pH112; lactose such as lactose monohydrate, lactose anhydrous and Pharmatose DCL21; dibasic calcium phosphate such as Emeompress® (JRS Pharma, Patterson, N.Y.); mannitol; starch; sorbitol; sucrose; and glucose.
  • the inert filler comprises microcrystalline cellulose.
  • the inert filler comprises a lactose selected from the group consisting of lactose monohydrate and lactose anhydrous.
  • the inert filler comprises a saccharide selected from the group consisting of mannitol, starch, sorbitol, sucrose, and glucose.
  • the saccharide is sorbitol.
  • the composition of the present invention may further comprise lubricants.
  • Any suitable lubricant may be used in the present invention.
  • the lubricant comprises agents that act on the flowability of the powder to be compressed.
  • Exemplary lubricants include, but are not limited to, colloidal silicon dioxide such as AerosilTM 200, talc, stearic acid, magnesium stearate, and calcium stearate.
  • the lubricant is stearic acid.
  • the composition of the present invention may further comprise disintegrants.
  • Any suitable disintegrant may be used in the present invention.
  • Exemplary disintegrants include, but are not limited to, lightly cross-linked polyvinyl pyrrolidone, corn starch, potato starch, maize starch and modified starches, croscarmellose sodium, cross-povidone, sodium starch glycolate and combinations and mixtures thereof.
  • the disintegrant is chosen from crospovidone and polyvinylpyrrolidone.
  • the composition described above may further comprise an enhancer.
  • the enhancer can be any suitable enhancer that is known to one of ordinary skill in the art.
  • Exemplary enhancers include, but are not limited to, a medium chain fatty acid salt, ester, ether or a derivative of a medium chain fatty acid which has a carbon chain length of from 4 to 20 carbon atoms.
  • the enhancer is solid at room temperature.
  • the enhancer is medium chain fatty acid salt, ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms.
  • the carbon chain length is from 8 to 14.
  • the enhancers are S-Cyclodextrins, vitamin E TPGS, gallic acid esters, crospovidones, sorbitan esters, poloxamers, or olyoxyethylene glycolated natural or hydrogenated castor oil (Cremophor®, BASF).
  • the enhancers are medium chain glycerides or a mixture of medium chain glycerides. Exemplary enhancers are further described in U.S. Patent Publication No. 2003/0091623, and U.S. Pat. No. 6,372,728 which are incorporated by reference in their entireties.
  • the GnRH related compound described herein may be prepared in the presence of a co-solvent system in a manner that the gelation of the GnRH compound is reduced.
  • the final step of the preparation of the GnRH related compound is conducted in the presence of a co-solvent system.
  • the final step is drying, using known techniques, such as lyophilization, tray drying, spray drying, fluid bed drying, or other similar techniques known to one skilled in the art.
  • the co-solvent system comprises water and at least one water-miscible solvent.
  • the water miscible solvent can be chosen from, but are not limited to, linear or branched C 1-6 alcohol, tetrahydrofuran, acetone, ethyl methyl ketone, methyl isobutyl ketone, methyl isopropyl ketone, cyclohexanone, diethyl ketone, pentan-3-one, cyclohexane, acetonitrile, N,N-dimethylformamide, N,N-dimethylacetamide, dioxane, alcohol, ethylene glycol, diglyme, monoglyme, ethylene glycol monomethyl ether, diethylene glycol, triethylene glycol, polyethylene glycol and a mixture thereof.
  • At least one water miscible solvent is a linear or branched C 1-6 alcohol.
  • exemplary suitable alcohols include, but are not limited to, methanol, ethanol, propanol, iso-propanol, butanol, sec-butanol, iso-butanol, tert-butanol, 1-pentanol, 2-pentanol, and hexanol.
  • at least one water miscible solvent is selected from the group consisting of methanol, ethanol, n-propanol, iso-propanol and tert-butanol.
  • At least one water miscible solvent is selected from the group consisting of methanol, ethanol, iso-propanol, tert-butanol, acetoniltrile and methylene chloride.
  • the present invention may be carried out with two or more water miscible solvents.
  • the weight ratio of the water miscible solvent to water is in the range of about 1/1000 to about 99/1. In some embodiments, the weight ratio is about 3/97 to about 59/41.
  • the process comprises further adding acid during the preparation of the GnRH related compound.
  • Acids used in the present invention include, but are not limited to acetic acid, sulfuric acid, hydrochloride acid, trifluoracetate, citrate acid, tartaric acid, ascorbic acid, and boric acid, etc.
  • the concentration of the acid is sufficient to prepare a salt of the GnRH related compound.
  • the concentration of the acid depends on the molecular weight of the GnRH related compound and the acid used herein. In some embodiments, the concentration is in the range of about 0.5% to about 20% of the solution.
  • the method comprises mixing the peptide, polypeptide, protein, an analog or derivative thereof with a sufficient amount of at least one stabilizing agents to improve the stability of peptide, polypeptide, protein, an analog and/or derivative thereof.
  • at least one stabilizing agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent.
  • the term “mixing” means contacting, combining, reacting, and/or coating a peptide, polypeptide, protein, an analog and/or derivative thereof, with one or more stabilizing agents.
  • “mixing” means combining a peptide, polypeptide, protein, an analog or derivative thereof with a stabilizing agent in a composition.
  • the GnRH related compound is prepared in the presence of a co-solvent system described herein.
  • the concentration of the stabilizing agent in the composition is equal to or above the critical micelle concentration of the stabilizing agent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of one or more peptide, polypeptide, protein, an analog or derivative thereof, and a sufficient amount of at least one stabilizing agents to improve the stability of the peptide, polypeptide, protein, an analog or derivative thereof, wherein at least one stabilizing agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to any substance, not itself a therapeutic agent, used as a vehicle for delivery of a therapeutic agent to a subject.
  • compositions of the present invention may be suitable for formulation for oral, parenteral, inhalation spray, topical, rectal, nasal, sublingual, buccal, vaginal or implanted reservoir administration, etc.
  • the compositions are administered orally, topically, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • the composition used in the present invention is in an oral dosage form.
  • the oral dosage form is chosen from tablets, capsules, granules, powders, capsules filled with granules or powders, capsules filled with liquids or semi-solids, sachets filled with granules or powders, liquid, emulsions, microemulsions, and any composition that capable of forming emulsions.
  • the oral dosage form may be a tablet, a multiparticulate, or a capsule.
  • the medium chain fatty acid and derivatives thereof described herein may also function as an enhancer.
  • the oral dosage form is a delayed release dosage form which minimizes the release of the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agents and/or the enhancer in the stomach, and hence the dilution of the local stabilizing agent or the concentration of the enhancer therein, and releases the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent or the enhancer in the intestine.
  • the oral dosage form is a delayed release rapid onset dosage form.
  • Such a dosage form minimizes the release of peptide, polypeptide, protein, an analog or derivative thereof and stabilizing agent or enhancer in the stomach, and hence the dilution of the local stabilizing agent or enhancer concentration therein, but releases the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent or the enhancer rapidly once the appropriate site in the intestine has been reached, maximizing the delivery of the poorly permeable or soluble peptide, polypeptide, protein, an analog or derivative thereof by maximizing the local concentration of the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent at the site of absorption.
  • a controlled release coating may be applied to the final dosage form (capsule, tablet, multilayer tablet etc.).
  • the controlled release coating may comprise a rate controlling polymer material described herein.
  • the dissolution characteristics of such a coating material may be pH dependent or independent of pH.
  • the composition of the present invention may have an enteric coating thereon.
  • the enteric coating comprises a polymer selected from the group consisting of poly(acrylic acid), polyacrylate, poly(methacrylic acid), polymethacrylate, and mixtures thereof.
  • the enteric coated composition may be in the form of a tablet or capsule.
  • tablette as used herein includes, but is not limited to, immediate release (IR) tablets, sustained release (SR) tablets, matrix tablets, multilayer tablets, multilayer matrix tablets, extended release tablets, delayed release tablets and pulsed release tablets any or all of which may optionally be coated with one or more coating materials, including polymer coating materials, such as enteric coatings, rate-controlling coatings, semi-permeable coatings and the like.
  • IR immediate release
  • SR sustained release
  • matrix tablets such as enteric coatings, rate-controlling coatings, semi-permeable coatings and the like.
  • extended release tablets delayed release tablets and pulsed release tablets any or all of which may optionally be coated with one or more coating materials, including polymer coating materials, such as enteric coatings, rate-controlling coatings, semi-permeable coatings and the like.
  • coating materials including polymer coating materials, such as enteric coatings, rate-controlling coatings, semi-permeable coatings and the like.
  • tablette also includes osmotic delivery systems in which a drug compound
  • Tablet solid oral dosage forms may be useful in the practice of the invention include those selected from the group consisting of IR tablets, SR tablets, coated IR tablets, matrix tablets, coated matrix tablets, multilayer tablets, coated multilayer tablets, multilayer matrix tablets and coated multilayer matrix tablets.
  • a tablet dosage form is an enteric-coated tablet dosage form.
  • a tablet dosage form is an enteric-coated rapid onset tablet dosage form.
  • Capsule solid oral dosage forms may be useful in the practice of the present invention include those selected from the group consisting of instant release capsules, sustained release capsules, coated instant release capsules, and coated sustained release capsules including delayed release capsules.
  • Capsules may be filled with powders, granules, multi particulates, tablets, semi-solids, or liquids.
  • a capsule dosage form is an enteric-coated capsule dosage form.
  • a capsule dosage form is an enteric-coated rapid onset capsule dosage form.
  • Capsules may be made of hard gelatin, soft gelatin, starch, cellulose polymers, or other materials as known to the art.
  • multiparticulate means a plurality of discrete particles, pellets, mini-tablets and mixtures or combinations thereof. If the oral form is a multiparticulate capsule, such hard or soft gelatin capsules can suitably be used to contain the multiparticulate. In some embodiments, a sachet may suitably be used to contain the multiparticulate. In some embodiments, the multiparticulate may be coated with a layer containing rate controlling polymer material. In some embodiments, a multiparticulate oral dosage form according to the invention may comprise a blend of two or more populations of particles, pellets, or mini-tablets having different in vitro and/or in vivo release characteristics. For example, a multiparticulate oral dosage form may comprise a blend of an instant release component and a delayed release component contained in a suitable capsule.
  • the multiparticulate and one or more auxiliary excipient materials can be compressed into tablet form such as a multilayer tablet.
  • a multilayer tablet may comprise two layers containing the same or different levels of the same active ingredient having the same or different release characteristics.
  • a multilayer tablet may contain different active ingredient in each layer.
  • Such a tablet, either single layered or multilayered, can optionally be coated with a controlled release polymer so as to provide additional controlled release properties.
  • multiparticulate dosage form comprises a capsule containing delayed release rapid onset minitablets.
  • a multiparticulate dosage form comprises a delayed release capsule comprising instant release minitablets.
  • a multiparticulate dosage form comprises a capsule comprising delayed release granules.
  • a multiparticulate dosage form comprises a delayed release capsule comprising instant release granules.
  • emulsion used herein means a suspension or dispersion of one liquid within a second immiscible liquid.
  • the emulsion is an oil-in-water or water-in-oil-in-water emulsion.
  • microemulsion used herein means a solution in which the hydrophobic (oil-like) phase and the hydrophilic (water-like) phase and a surface active agent form micelle structures. Such dispersions are clear and stable over time.
  • emulsion or “microemulsion”, used herein includes a hydrophilic or a hydrophobic liquid which, on dilution with a hydrophobic or a hydrophilic liquid respectively, form an emulsion or a microemulsion.
  • emulsion or ‘microemulsion’, used herein may include solid or semi-solid materials which may be liquid at higher temperatures.
  • the material may be solid at room temperature. At about body temperature (about 37° C.), the material may be liquid.
  • compositions of this invention may be in the form of a suppository for rectal administration.
  • the suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, beeswax and polyethylene glycols.
  • compositions of the present invention may be in the form of a topical solution, ointment, or cream in which the active component is suspended or dissolved in one or more carriers.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2 octyldodecanol, benzyl alcohol and water.
  • the pharmaceutically acceptable composition when it is an ophthalmic formulation, it may be a micronized suspension in isotonic, pH adjusted sterile aqueous solution, or as a solution in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in the form of an ointment.
  • compositions of this invention may also be administered by nasal, aerosol or by inhalation administration routes.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, gender, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • Another aspect of the present invention provides a method of treatment of a medical condition treatable by a peptide, polypeptide, protein, an analog or derivative thereof comprising administering to a patient suffering from said condition a pharmaceutical composition as described in the present application.
  • the medical condition includes, but is not limited to, diabetes mellitus, Alzheimer, autoimmune diseases, colon cancer, sex hormone dependent disease such as benign prostate hyperplasia, prostate cancer, estrogen-dependent breast cancer, endometrial cancer, ovarian cancer, endometriosis and precocious puberty, and contraception in a human or animal subject
  • compositions as described in the present application in the manufacture of a medicament for the treatment of a medical condition treatable by said peptide, polypeptide, protein, analogs and/or derivatives thereof.
  • FIG. 1 The six different acyline batches are described in FIG. 1 , which summarizes the variations in the lyophilization procedure, which is the last step of the preparation of acyline.
  • a co-solvent system is used in the lyophilization step. Water is used as a solvent for other batches.
  • the batches and the associated lyophilization solvents are also illustrated in FIG. 1 .
  • the acyline may be prepared according to methods known to one of skill in the art, for example U.S. Pat. No. 6,747,125, or processes described in the present application.
  • the study of gelation for different acyline batches in propylene glycol is carried out and the result is summarized in FIG. 2 .
  • the 9% and 16.7% of acyline sampls are prepared by using the acyline batches listed in FIG. 1 .
  • the 9% acyline sample of XF 173/315-125, XF 185/165-133 and XF 173/315-151A are prepared by using acyline prepared via use of a co-solvent during the lyophilization step.
  • the acyline batches of XF 173/315-125, XF 185/165-133 and XF 173/315-151A appear clear and non-viscous after 2 hours. Other batches which are lyophilized by using water as a solvent did not appear as clear and non-viscous solutions due to the presence of gelled acyline.
  • the gelation of different acyline drug batches in water is investigated and the results are summarized in FIG. 3 .
  • the 0.5% and 1% acyline samples are prepared by using the acyline batches listed in FIG. 1 .
  • the 0.5% and 1% of acyline samples of XF 173/315-125, XF 185/165-133 and XF 173/315-151A which are prepared by using co-solvent during the lyophilization step appear clear and non viscous upon dissolving in water.
  • FIG. 4( a ) The formulation of the standard micro emulsion is shown in FIG. 4( b ).
  • the 5 mg and 10 mg formulations of acyline in a microemulsion are prepared by using the acyline lots synthesized in FIG. 1 .
  • 5 mg and 10 mg dose of acyline batches of XF 173/315-151A which are prepared by using co-solvent during the lyophilization step appeared clear and transparent after 2 hours.
  • acyline composition Different concentrations of acyline composition are prepared by adding acyline to pH 6.8 buffer solution either at room temperature or 37° C. All acyline solutions contain 0.6 mg/mL sodium caprate (sodium caprate is referred as C 10 in the figures). All samples are centrifuged and filtered prior to analysis. Samples are analyzed by reverse phase HPLC with UV detection.
  • acyline 10 mg is transferred into 200 mL volumetric flask.
  • a pH 6.8 buffer solution is preheated to 37° C.
  • 100 mL pre-heated buffer solution and 0.6 mg/mL sodium caprate is added to the flask to prepare a 0.1 mg/mL acyline sample.
  • Tween 80 is added to prepare a 0.1 mg/mL acyline solution with 1% Tween 80.
  • Samples are shaken in a temperature controlled water bath at 37° C. 5 mL samples are taken at 1, 5, 10, 15, 20, 30 and 120 minutes after mixing acyline with the buffer solution. Samples are filtered immediately through 0.45 ⁇ m filters and the first 3 mL is discarded.
  • acyline sample for example 5.
  • the result of comparison of 5 mg/mL, 1.0 mg/mL, 0.1 mg/mL, and 0.1% Tween in phosphate buffer containing 0.6 mg/mL of sodium caprate and 0.1 mg/mL acyline is graphically recorded in FIG. 6 .
  • the results show that only 1% Tween 80 solution may completely inhibit the gelation of acyline.
  • acyline sample for example 6.
  • the result of comparison of 1% and 1 mg/mL Tween 80 in 0.1 and 0.01 mg/mL acyline sample is graphically recorded in FIG. 7 .
  • the results show that 1% Tween 80 reduces gelation for both 0.1 mg/mL and 0.01 mg/mL acyline sample.
  • acyline sample for example 7 A similar experimental procedure as Example 4 is used to prepare acyline sample for example 7.
  • the result of comparison of 0.01 mg/mL acyline samples having 0.1%, 0.5% and 1% Tween 80 is graphically recorded in FIG. 8 .
  • the results show that the acyline sample with 1% Tween 80 significantly reduces the gelation.
  • FIG. 9 graphically demonstrates the impact of different concentrations of sodium caprate (C10) on the gelation of acyline in water.
  • concentration of sodium caprate is below 10 mg/mL
  • the recovery of acyline significantly decreases, which implies an increase of gelation of acyline.
  • the investigators of the present invention believe that the increase of the gelation is due to an increased concentration of ions caused by the addition of sodium caprate.
  • concentration of sodium caprate reaches and is above the CMC of sodium caprate ( ⁇ 20 mg/mL)
  • the CMC of sodium caprate is known as 100 mM ( ⁇ 20 mg/mL).
  • FIG. 11 graphically demonstrates the relative bioavailability of various formulations of acyline in dogs.
  • the relative bioavailability is measured by comparing the absolute bioavailability of various formulations of acyline with the absolute bioavailability of a standard formulation of acyline, which is a formulation without any anti-gelling agent.
  • the formulation of microemulsion 1 (M 1 /55% Capmul MCM), microemulsion 2 (M 2 /45% Capmul PG-8), and microemulsion 3 (M 3 /55% Capmul MCM C10) are illustrated in FIG. 10( a ) through (c).
  • the formulation of the standard solution is 5 mg acyline, 550 mg sodium caprate and 5 mL purified water. “C 10 ” in FIGS.
  • FIG. 12 graphically demonstrates the relative bioavailability of (1) enteric tablets of 10 mg acyline versus unenhanced 5 mg acyline solution sample and (2) enteric tablets of 10 mg acyline versus 5 mg acyline standard sample.
  • the formulation of acyline standard sample is 5 mg acyline, 550 mg sodium caprate and 5 mL purified water.
  • the acyline in the tablets and the 5 mg acyline sample is prepared by using a lyophilization step with water as the solvent.
  • the tablets contained the same amount of sodium caprate as the 5 mg acyline sample. It is observed that, the concentration of sodium caprate, 110 mg/mL, is sufficient to reduce or even inhibit gelation in the solution and enhance the bioavailability of acyline solution.
  • FIG. 12 demonstrates that the bioavailability of enhanced tablets is significantly improved compared to unenhanced tablet.
  • the samples are filtered (Millex-HV 0.45 ⁇ m), and the first 5 ml of filtrate discarded.
  • the appropriate test solutions are stirred and additional samples are collected from all test solutions at 6, 24 hours etc.
  • the amount of bovine insulin in solution after filtration is determined against standards (70 mg in 200 ml of 0.01 N HCl).
  • the % recovery of intact insulin after the insulin solution stands for a period of time is recorded in Tables 2 and 3 and graphically illustrated in FIGS. 13( a ) and ( b ). It is observed that insulin solutions with sodium caprate are significantly more stable than the insulin samples without sodium caprate or the solutions with capric acid. For insulin samples that contain sodium caprate, at least 90% of insulin is recovered after the solutions stands at 37° C. for at least about 24 hours.

Abstract

The present invention provides composition of comprising a therapeutically effective amount of at least one peptide, polypeptide, analog or derivative thereof and a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, an analog or derivative thereof, wherein at least one stabilizing agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent. The method for preparation of a composition of a peptide, polypeptide, protein, an analog and/or derivative thereof is also provided. The process comprises mixing the peptide, polypeptide, protein, an analog or derivative thereof with a sufficient amount of at least one stabilizing agents to improve the stability of the peptide, polypeptide, protein, an analog or derivative thereof, and the agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent.

Description

    RELATED APPLICATIONS
  • This application claims the benefit under 35 U.S.C. §119(e) of U.S. Provisional Patent Application Ser. No. 61/051,038, filed May 7, 2008, the disclosures of which is incorporated herein by reference in its entirety.
  • FIELD OF THE INVENTION
  • The present invention generally relates to compositions of peptides, polypeptides, proteins, analogues or derivatives thereof, and the process of preparation thereof.
  • BACKGROUND OF THE INVENTION
  • A variety of biologically active peptides, polypeptides and proteins have been widely used for medical treatment. For a majority of medical treatments, there is a need to deliver a sustained level of biologically active peptides or proteins to animals or humans to provide a stable therapeutic effect. Additionally, the biologically active peptides or proteins also need to be stored for a prolonged period of time and still maintain their activity. However, many naturally occurring and synthetic peptides and proteins as well as their analogs have exhibited a tendency to form gels, aggregates, fibrils, dimers, other polymers, coagulates, etc. In some cases, the formed material may be able to revert back to the active monomer form. In other cases, the altered state may be permanent and represent a degraded state. Some exemplary biologically active peptides include insulin, glucagon-like peptide-1 (GLP-1) and Gonadotropin-releasing hormone (GnRH) analogs. The instability of the peptides has become a significant barrier for the preparation, process, storage and delivery of these peptides.
  • To date, a few methods have been discovered that may be used to stabilize solution of peptides or proteins. For example, U.S. Pat. No. 6,124,261 describes a non aqueous polar aprotic peptide formulation that may be used to stabilize the peptide. In another example, a citrate buffering agent may be used to reduce the gelation of fatty acid-acylated protein. See U.S. Pat. No. 5,631,347.
  • However, due to the diversity of biologically active peptides and proteins, there is a continuing need to find new compositions of peptides or proteins with improved stability and processes of preparing compositions of peptides or proteins.
  • SUMMARY OF THE INVENTION
  • The present invention provides compositions comprising a therapeutically effective amount of at least one peptide, polypeptide, protein, an analog or derivative thereof and a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof in a solution, wherein at least one stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent. In some embodiments, at least one peptide, polypeptide, protein, an analog or derivative thereof is insulin, or an analog or derivative thereof. In one embodiment, at least one peptide, polypeptide, protein, analog or derivative thereof is Glucagon-Like Peptide (GLP), or an analog or derivatives thereof. In another embodiment, the stabilizing agent is selected from the group consisting of sodium caprylate, sodium caprate and sodium laurate.
  • According to another aspect of the present invention, methods for preparation of a composition of at least one peptide, polypeptide, protein, analog or derivative thereof are provided. In some embodiments, the methods comprises mixing the peptide, polypeptide, protein, analog or derivative thereof with a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof, and the stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms. In one embodiment, the stabilizing agent is a medium chain fatty acid salt has a carbon chain length of from about 8 to about 14 carbon atoms.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.
  • FIG. 1 is a table describing the preparation of different acyline batches.
  • FIG. 2 shows appearance of 9% and 16.7% acyline samples of different acyline batches in propylene glycol.
  • FIG. 3 shows the appearance of 0.5% and 1% acyline samples of different acyline batches in water.
  • FIG. 4( a) shows the appearance of 5 mg and 10 mg doses acyline microemulsions of different acyline batches.
  • FIG. 4( b) shows the formulation of the microemulsion.
  • FIG. 5 graphically demonstrates the comparison results of the gelation of 0.1 mg/mL acyline sample with 1% and 0% Tween 80.
  • FIG. 6 graphically demonstrates the comparison results of the gelation of 0.1 mg/mL acyline sample having 5 mg/mL, 1 mg/mL, 0.1 mg/mL and 1% Tween 80.
  • FIG. 7 graphically demonstrates the comparison results of the gelation of 0.1 and 0.01 mg/mL acyline sample having 1% and 1 mg/mL Tween 80.
  • FIG. 8 graphically demonstrates the comparison results of the gelation of 0.01 mg/mL acyline sample having 0.1%, 0.5% and 1% Tween 80.
  • FIG. 9 graphically demonstrates the correlation between the concentration of sodium caprate and the gelation of acyline.
  • FIG. 10( a) illustrates the formulation of microemulsion 1—55% Capmul MCM. FIG. 10( b) illustrates the formulation of microemulsion 2—45% Capmul PG-8. FIG. 10( c) illustrates the formulation of microemulsion 3—55% Capmul MCM C10.
  • FIG. 11 graphically demonstrates the comparison results of relative bioavailability of different formulations of acyline.
  • FIG. 12 graphically demonstrates the comparison results of relative bioavailability of (1) enteric tablets 10 mg acyline versus acyline sample with no surface active agent and (2) enteric tablets 10 mg acyline versus 5 mg acyline with sodium caprate sample.
  • FIG. 13( a) illustrates the correlation of % of recovery of insulin for insulin formulations with or without sodium caprate. FIG. 13( b) illustrates % of recovery of insulin from insulin formulations with capric acid.
  • DETAILED DESCRIPTION
  • The foregoing and other aspects of the present invention will now be described in more detail with respect to the description and methodologies provided herein. It should be appreciated that the invention can be embodied in different forms and should not be construed as limited to the embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the invention to those skilled in the art.
  • All patents, patent applications and publications referred to herein are incorporated by reference in their entirety. In case of a conflict in terminology, the present specification is controlling.
  • The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used in the description of the embodiments of the invention and the appended claims, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. Also, as used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items. Furthermore, the term “about,” as used herein when referring to a measurable value such as an amount of a compound, dose, time, temperature, and the like, is meant to encompass variations of 20%, 10%, 5%, 1%, 0.5%, or even 0.1% of the specified amount. Unless otherwise defined, all terms, including technical and scientific terms used in the description, have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
  • As used herein, “alcohol” means an organic compound in which one or more hydroxyl (OH) groups are attached to carbon (C) atoms in place of hydrogen (H) atoms. In some embodiments, the alcohol contains 1-6 carbon atoms. Yet, in other embodiments, the alcohol contains 1-4 carbon atoms. Exemplary alcohols include, but are not limited to, methanol, ethanol, n-propanol, iso-propanol, butanol, tert-butanol, pentanol, hexanol.
  • As used herein, “gelation” means that a compound of interest undergoes aggregation to form fibrils, dimers, longer polymers, coagulates, or structures that may result in formation of a colloid structure or gel. The viscosity of the mixture, used in the present application, may be applied to a compound of interest in aqueous solution or a solid mass.
  • “Gelation” may occur to varying extents, and may occur in such a maimer as to be non-detectable by ordinary means. For example, there may be no increased viscosity or changes in the flow characteristics of the solution. However, the material formed from the gelation (hereinafter “gel”) may be removed by physical means such as filtration and thus detected by analytical techniques known to one skilled in the art. The presence of gels may cause significant problems in the development of different administration forms. For example, when the gelled system is processed to obtain powders of the drug as part of drug substance manufacturing techniques, a xerogel may be formed during the process. As used herein, ‘xerogel’ is a solid formed from the gel after the liquid is removed from a gelled system. The powder obtained from the gelled system containing xerogel may have substantially different characteristics from powders obtained from solutions that contain no gelled material. In some situations, obtained power loses the biological activity. Therefore, the formation of gels may potentially cause deleterious effects at either the drug substance processing stage, or at the stage of preparing the dosage form or storage stage.
  • The degree of agglomeration or coalescence which results in the formation of the gel may be measured by various methods known to one skilled in the art, for example micro or macro filtration followed by assay of the filtrate, centrifugation followed by assay of the supernatant, or various optical absorption or diffraction methods including those utilizing visible or UV light methods, or laser based methods. The methods of measuring physical characteristics of the liquid may also be used such as surface tension, freezing point depression, viscosity, or measurement of other colligative properties. The degree of xerogel formation may be measured by dispersing the material in water and using the techniques described herein or by direct measurement on the powder by methods known to one skilled in the art including x-ray powder diffraction, IR spectroscopy, or other methods known to the art that may be carried out on powdered materials.
  • As used herein, the term “reduce the gelation” means to disrupt, retard or eliminate the gel formation of a compound. In situation where the gelation is reversible, the term “reduce the gelation” also means to reverse the gel back to monomeric form of the compound, which maintains it biological activity. As used herein, the term “anti-gelling agent” refers to an agent, a compound, a composition or a combination thereof which may inhibit the gelation of at least 50% of the compound of interest, when a sufficient amount of the anti-gelling agent is used. In some embodiments, the anti-gelling agent may inhibit the gelation of at least 80% of the compound of interest, when a sufficient amount of the anti-gelling agent is used. In some embodiments, the anti-gelling may inhibit the gelation of at least 90% of the compound of interest, when a sufficient amount of the anti-gelling agent is used.
  • As used herein, a “therapeutically effective” or “therapeutically acceptable” amount refers to an amount that will elicit a therapeutically useful response in a subject. The therapeutically useful response may provide some alleviation, mitigation, or decrease in at least one clinical symptom in the subject. Those skilled in the art will appreciate that the therapeutic useful response need not be complete or curative, as long as some benefit is provided to the subject. In some embodiments, the subject is an animal. In some embodiments, the subject is a human.
  • As used herein, the term “water miscible solvent” means a solvent that can be mixed with water to form a solution. Water miscible solvents may be used to create a hydrophilic phase.
  • The present invention provides compositions comprising, consisting essentially of, or consisting of, a therapeutically effective amount of at least one peptide, polypeptide, protein, an analog or derivative thereof and a sufficient amount of at least one stabilizing agents to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof. As used herein, a “stabilizing agent” is an agent that improves the stability of a peptide, polypeptide, protein, an analog or derivative thereof described herein.
  • In some embodiments, at least one stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms. In some embodiments, at least one stabilizing agent is a surface active agent.
  • The present invention provides compositions that maintain the stability of peptide, polypeptide, protein, an analog or derivative thereof described herein for a sufficient time to be stored, processed and/or administered for treatment. One way of measuring the stability is measuring the percentage of peptide, polypeptide, protein, an analog or derivative thereof is retained under a specific condition. As used herein, the term “peptide, polypeptide, protein, analog or derivative thereof is retained” means that the peptide, polypeptide, protein, an analogs or derivative thereof does not form gels, aggregates, fibrils, dimers, other polymers, coagulates, rather it maintains as a monomer or its biological activity after a period of time. In one embodiment, in the composition of the present invention, at least about 50% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, in the composition of the present invention, at least about 80% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, in the composition of the present invention, at least about 90% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, in the composition of the present invention, at least about 95% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, the stability is improved when the gelation of peptide, polypeptide, protein, an analog or derivative thereof is reduced.
  • The compositions described herein may be used directly for storage, processing or administration. Alternatively, the composition may also be used to mix with a suitable medium (e.g. a solvent, a microemulsion or a solid mass) for storage, processing or administration. In the situations where a medium is used, the stability of peptide, polypeptide, protein, analogues or derivatives thereof in the medium is improved as described herein.
  • In some embodiments, the stabilizing agent is an anti-gelling agent. In one embodiment, the present invention provides compositions comprising, a therapeutically effective amount of one or more peptide, polypeptide, protein, an analog or derivative thereof, and a sufficient amount of at least one anti-gelling agents to reduce the gelation of the peptide, polypeptide, protein, an analog or derivative thereof.
  • In one embodiment, the dissolution rate of the stabilizing agent and the peptide, polypeptide, protein, analogs or derivatives thereof in the composition are substantially the same.
  • I. Peptide, Polypeptide, Protein, an Analog and Derivative Thereof
  • The present invention can be applied to any peptide, polypeptide, protein, analog or derivative that has a tendency to aggregates, dimerizes, polymerizes, coagulates, gels or fibrillates. Analogs, derivatives, and pharmaceutically acceptable salts of any of the peptides, polypeptides or proteins are included in these terms. As used herein, the “tendency to to aggregates, dimerize, polymerize, coagulate, gel or fibrillate” refers to that at least 50% of a compound of interest undergoes aggregation to form fibrils, dimers, polymers that coagulates, or structures that may result in formation of a colloid structure or gel in a system at a certain temperature (e.g. 37° C.) after the system stands for a period of time (e.g. at least about 24 hours).
  • In some embodiments, at least one peptide, polypeptide, protein, an analog or derivative thereof is an insulin or an analog or derivative thereof.
  • In one embodiment, at least one peptide, polypeptide, protein, analogs or derivatives thereof is Glucagon-Like Peptide (GLP), or an analog or derivative thereof. In another embodiment, at least one GLP is selected from the group consisting of GLP-1, GLP-1 analogs, derivatives thereof, GLP-2, GLP-2 analogs, derivatives thereof, and exendin-4, analogs and derivatives thereof.
  • More exemplary peptides, polypeptides, proteins, analogs or derivatives thereof include, but are not limited to, GnRH agonists and antagonists, somatostatin, ACTH, corticotropin-releasing factor, angiotensin, calcitonin, gastric inhibitory peptide, growth hormone, growth hormone-releasing factor, pituitary adenylate, exendin, exendin-3, cyclase activating peptide, secretin, enterogastrin, somatostatin, somatotropin, somatomedin, parathyroid hormone, thrombopoietin, erythropoietin, hypothalamic releasing factors, prolactin, thyroid stimulating hormones, endorphins, enkephalins, vasopressin, oxytocin, opioids and analogues thereof, superoxide dismutase, interferon, asparaginase, arginase, arginine deaminase, adenosine deaminase, ribonuclease, FVII, FXIII, a mixture of FVII and FXIII, IL-20, IL-21, IL-28a, IL-29, IL-31, analogs and derivatives thereof.
  • In some embodiments, at least one peptide, polypeptide, protein, analog or derivative comprises a variety of GnRH related compounds which have a tendency of gelation. As used herein, the “tendency of gelation” refers to that at least 50% of a compound undergoes aggregation to form fibrils, dimers, polymers that coagulates, or structures that may result in formation of a colloid structure or gel in a system at a certain temperature (e.g. 37° C.) after the system stands for a period of time (e.g. at least about 2 hours). As used herein, GnRH related compounds include both GnRH antagonists and GnRH agonists. In some embodiments, the present invention may be applied to GnRH antagonists. In some embodiments, the present invention include, but is not limited to, the following GnRH antagonists, acyline (Ac-D2Nal-D4Cpa-D3Pal-Ser4Aph(Ac)-D4Aph(Ac)-Leu-ILys-Pro-DAla-NH2), Acetyl-β-[2-Naphthyl]-D-Ala-D-p-Chloro-Phe-β-[3-Pyridyl]-D-Ala-Ser-Nε-[Nicotinoyl]-Lys-Nε-[Nicotinoyl]-D-Lys-Leu-Nε-[Isopropyl]-Lys-Pro-D-Ala-NH2 (also referred to herein as Antide), acetyl-D2Nal1, D4CIPhe2, D3Pal3, ARg5, Dglu6 (AA) (also referred to herein as NalGlu), acetyl-D2Nal-D4CIPhe-D3Pal-Ser-Aph(Ac)-D-Aph(Ac)-Leu-Lys(lpr)-Pro-D-Ala-NH2, Abarelix (Specialty European Pharma, Dusseldorf, Germany), Nal-Lys, Synarel, (Searle Peapack, N.J.), Ganirelix (Orgalutron/Antagon) (Organan, West Orange, N.J.), Cetrorelix I (Aeterna Zentaris Inc, Frankfurt, Germany), Cetrotide, Azaline B, new generation long-acting GnRH analogues incorporating p-ureido-phenylalanines at positions 5 and 6 (such as Degarelix), FE200486, Ac-D2Nal-D4Cpa-D3Pal-Ser-4Aph(L-hydroorotyl)-D4Aph(carbarnoyl)-Leu-ILys-Pro-DAla-NH2 (the acetate salt of which is FE200486), Ac-D2Nal-D4Cpa-D3Pal-Ser-4Aph(Atz)-D4Aph(Atz)-Leu-ILys-Pro-DAla-NH2 wherein Atz is 3′-amino-1H-1′,2′,4′-triazol-5′-yl, and the antagonists described in U.S. Pat. Nos. 5,506,207, 5,821,230, 5,998,432, 6,156,772, 6,156,767, 6,150,522, 6,150,352, 6,147,088, 6,077,858, 6,077,847, 6,025,366, 6,017,944, 6,004,984, 6,214,798, and 6,875,843. In some embodiments, the GnRH antagonists of the present invention have a tendency of gelation in the presence of ions. In some embodiments, at least one GnRH antagonist is selected from the group consisting of acyline, abarelix, azaline B, cetrorelix, ganirelix, teverelix, degarelix, antide, orntide and GnRH antagonists described in U.S. Pat. No. 7,098,305.
  • In one embodiment, at least one GnRH antagonist is selected from the group consisting of abarelix, cetrorelix, degarelix, ganirelix, and a pharmaceutically acceptable salt thereof.
  • As used throughout this specification and claims, the term “abarelix” refers to a compound having a structure of Formula I
  • Figure US20090280169A1-20091112-C00001
  • The IUPAC name of Formula I is acetyl-D-β-naphthylalanyl-D-4-chlorophenylalanyl-D-3-pyridylalanyl-L-seryl-L-N-methyl-tyrosyl-D-asparagyl-L-leucyl-L-N(e)-isopropyl-lysyl-L-prolyl-D-alanyl-amide. The term “abarelix” includes the compound of Formula I, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these. The term “abarelix” also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula I and pharmaceutically acceptable salts thereof.
  • As used throughout this specification and claims, the term “cetrorelix” refers to a compound having a structure of Formula II.
  • Figure US20090280169A1-20091112-C00002
  • The IUPAC name of Formula II is Acetyl-D-3-(2′-naphtyl)-alanine-D-4-chlorophenylalanine-D-3-(3′-pyridyl)-alanine-L-serine-L-tyrosine-D-citruline-L-leucine-L-arginine-L-proline-D-alanine-amide. The term “cetrorelix” includes the compound of Formula II, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these. The term “cetrorelix” also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula II and pharmaceutically acceptable salts thereof.
  • As used throughout this specification and claims, the term “degarelix” refers to a compound having a structure of Formula III.
  • Figure US20090280169A1-20091112-C00003
  • The IUPAC name of Formula III is N-acetyl-3-(naphtalen-2-yl)-D-alanyl-4-chloro-D-phenylalanyl-3-(pyridin-3-yl)-D-alanyl-L-seryl-4-((((4S)-2,6-dioxohexahydropyrimidin-4-yl)carbonyl)amino)-L-phenylalanyl-4-(carbamoylamino)-D-phenylalanyl-L-leucyl-N6-(1-methylethyl)-L-lysyl-L-prolyl-D-alaninamide. It is also known as FE-200486. The term “degarelix” includes the compound of Formula III, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these. The term “degarelix” also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula III and pharmaceutically acceptable salts thereof.
  • As used throughout this specification and claims, the term “ganirelix” refers to a compound having a structure of Formula IV.
  • Figure US20090280169A1-20091112-C00004
  • The IUPAC name of Formula IV is a (2S)-1-[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2R)-2-[[(2S)-2-[[(2R)-2-[[(2R)-2-[[(2R)-2-acetamido-3-naphthalen-2-ylpropanoyl]amino]-3-(4-chlorophenyl)propanoyl]amino]-3-pyridin-3-ylpropanoyl]amino]-3-hydroxypropanoyl]amino]-3-(4-hydroxyphenyl)propanoyl]amino]-6-[bis(ethylamino)methylideneamino]hexanoyl]amino]-4-methylpentanoyl]amino]-6-[bis(ethylamino)methylideneamino]hexanoyl]-N-[(2R)-1-amino-1-oxopropan-2-yl]pyrrolidine-2-carboxamide. The term “ganirelix” includes the compound of Formula IV, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these. The term “ganirelix” also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula IV and pharmaceutically acceptable salts thereof.
  • In some embodiments, the present invention may be applied to GnRH agonists that have a tendency of gelation. The exemplary GnRH agonists include, but are not limited to, histerelin, leuprolide and goserelin.
  • The terms “GnRH related compound”, “GnRH antagonist” and “GnRH agonist” include all forms thereof including stereoisomers, enantiomers, diastereomers, racemic mixtures, and derivatives thereof, for example, salts, acids, esters and the like. The compound may be provided in any suitable phase state including as solid, liquid, solution, suspension and the like. When provided in a solid particulate form, the particles may be of any suitable size or morphology and may assume one or more crystalline, semi-crystalline and/or amorphous forms.
  • The peptide, polypeptide, protein, analogs or derivatives there of used in the present invention may be present in any amount which is sufficient to elicit a therapeutic effect and, where applicable, may be present either substantially in the form of one optically pure enantiomer or as a mixture, racemic or otherwise, of enantiomers. As will be appreciated by those skilled in the art, the actual amount of peptide, polypeptide, protein, analogs or derivatives there of used in the composition will depend on the potency of the selected compound in question.
  • The peptides, polypeptides, proteins, analogs or derivatives described herein may be obtained through commercial resources or may be prepared according to methods known to one skill in the art. The GnRH antagonists applied in the present invention may be prepared using a method known to one of ordinary skill in the art or a process described in the present invention. For example, acyline can be prepared according to the method described in U.S. Pat. No. 5,506,207.
  • II. Stabilizing Agents
  • The amount of the stabilizing agent used in the present invention can vary considerably. For example, the amount can be dependent upon individual stabilizing agents, solvent systems and other components in the composition. Generally, the amount of stabilizing agent should be sufficient to improve the stability of peptide, polypeptide, protein, analogs or derivatives thereof in a system such that at least 50% of peptide, polypeptide, protein, an analog or derivative thereof is retained when the composition stands at 37° C. for at least about 24 hours. In some embodiments, the amount of stabilizing agent should be sufficient to improve the stability of peptide, polypeptide, protein, analogs or derivatives thereof in a system such that at least 80% of peptide, polypeptide, protein, analog or derivative thereof is retained when the composition stands at 37° C. for at least about 24 hours. In other embodiments, the amount of anti-gelling agent should be sufficient to be sufficient to improve the stability of peptide, polypeptide, protein, an analog or derivative thereof in a system such that at least 90% of peptide, polypeptide, protein, an analog or derivative thereof is retained when the composition stands at 37° C. for at least about 24 hours. In some embodiments, the concentration of the stabilizing agent in the composition is at least equal to or above the CMC (Critical Micelle Concentration) of the stabilizing agent in the composition.
  • A. Medium Chain Fatty Acid and Derivatives Thereof
  • In some embodiments, at least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 4 to 20 carbon atoms. In some embodiments, at least one stabilizing agent is medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms. In some embodiments, the carbon chain length is from 8 to 14. In some embodiments, at least one stabilizing agent is a salt of medium chain fatty acid and has a carbon chain length of from 8 to 14 carbon atoms. In some embodiments, at least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms; with the provisos that (i) where the stabilizing agent is an ester of a medium chain fatty acid, said chain length of from 6 to 20 carbon atoms relates to the chain length of the carboxylate moiety, and (ii) where the stabilizing agent is an ether of a medium chain fatty acid, at least one alkoxy group has a carbon chain length of from 6 to 20 carbon atoms. In another embodiment, at least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid which is solid at room temperature and which has a carbon chain length of from 8 to 14 carbon atoms; with the provisos that (i) where the stabilizing agent is an ester of a medium chain fatty acid, said chain length of from 8 to 14 carbon atoms relates to the chain length of the carboxylate moiety, and (ii) where the stabilizing agent is an ether of a medium chain fatty acid, at least one alkoxy group has a carbon chain length of from 8 to 14 carbon atoms. In some embodiments, at least one stabilizing agent is a sodium salt of a medium chain fatty acid, the medium chain fatty acid having a carbon chain length of from 8 to 14 carbon atoms. In some embodiments, the stabilizing agent is solid at room temperature. In another embodiment, at least one stabilizing agent is selected from the group consisting of sodium caprylate, sodium caprate, and sodium laurate. In one embodiment, at least one stabilizing agent is sodium caprate.
  • B. Surface Active Agents
  • In some embodiments, at least one stabilizing agent is a surface active agent. As used herein, the term “surface active agent” refers to an agent that lowers the surface tension of the medium in which it is dissolved and/or the interfacial tension with other phases, and, accordingly, is positively adsorbed at the liquid/vapor and/or at other interfaces. The surface active agents employed in the present invention include both ionic agents, i.e., cationic, anionic or zwitterionic, and non-ionic agents, or a mixture thereof.
  • Examples of cationic surface active agents include, but are not limited to, benzalkonium chloride, dicetyl ammonium chloride, cetyldimethylethylammonium bromide, cetylpyridinium chloride and salts of the above surface active agents.
  • Examples of anionic surface active agents include, but are not limited to, sodium stearoyl lactylate, hydrogenated lecithin, sodium lauryl sulfate, C8-32 fatty acids and salts thereof, cholic acid and derivatives thereof such as deoxycholate, and its salts, ursodeoxycholic acid, and taurocholic acid; C8-56 diesters of tartaric acid; phospholipids such as phosphatidic acid and phosphatidyl serine; C5-29 monoesters of lactic acid; C8-20 sulfonates, including alkyl-, olefin-, and alkylaryl derivatives; tridecyl- and dodecylbenzene sulfonic acids; and C5-33 sarcosine and betaine derivatives.
  • Examples of zwitterionic surface active agents include, but are not limited to, phospholipids such as lecithin, phosphatidylethanolamine, sphingomyelins, dodecyl betaine, dodecyl dimethylamine oxide, cocamidopropyl betaine, and coco ampho glycinate.
  • Examples of non-ionic surface active agents include, but are not limited to, steareths; polyethylene glycol (PEGs); polysorbates (e.g. Tween 80); cetearyl glucoside; various commercially available sorbitans and their derivatives, for example, sorbitan hexastearate ethoxylate EO 6 mole, sorbitan isostearate, sorbitan laurate, sorbitan monoisostearate ethoxylate EO 20 mole, sorbitan monolaurate ethoxylate EO 20 mole, sorbitan monooleate ethoxylate EO 20 mole, sorbitan monopalmitate ethoxylate EO 20 mole, sorbitan monostearate ethoxylate EO 20 mole, sorbitan monstearate ethoxylate EO 6 mole, Sorbitan oleate, sorbitan palmitate, sorbitan sesquioleate, sorbitan stearate, sorbitan tetraoleate ethoxylate EO 30 mole, sorbitan tetraoleate ethoxylate EO 40 mole, sorbitan tetraoleate ethoxylate EO 6 mole, sorbitan tetrastearate ethoxylate EO 60 mole, sorbitan trioleate ethoxylate EO 20 mole, sorbitan trioleate, sorbitan tristearate ethoxylate EO 20 mole, and sorbitan tristearate; ethoxylated castor oil, C5-29 mono-glycerides and ethoxylated derivatives thereof; C15-60 diglycerides and polyoxyethylene derivatives thereof having 1 to 90 POE groups; C10-40 esters (10-40 carbon atoms in the alcohol) of long chain fatty acids (fatty acids having 16 carbon atoms and above); C10-40 alcohols; sterols such as cholesterol, ergosterol, and C2-24 esters thereof; C8-96 ethoxylated fatty esters; C14-130 sucrose fatty esters; and polyoxyethylene (POE) derivatives thereof having 0 to 90 POE groups, e.g., polyoxyethylene sorbitan monooleate, sorbitol hexaoleate POE (50).
  • In some embodiments, at least one surface active agent is selected from the group consisting of sodium lauryl sulfate, polysorbate surface active agents (such as polysorbate 20 (Tween 20), polysorbate 80 (Tween 80)), sorbitan surface active agents, sorbitan monolaurate and polyethoxylated castor oil and a combination thereof. In some embodiments, at least one surface active agent comprises polysorbate.
  • III. Additional Excipients
  • According to some aspects of the present invention, the composition of the present invention further comprises one or more excipients. As will be appreciated by those skilled in the art, the exact choice of excipients and their relative amounts will depend to some extent on the final dosage form. In some embodiments, the excipients are selected from the group consisting of rate-controlling polymeric materials, diluents, lubricants, disintegrants, plasticizers, anti-tack agents, opacifying agents, pigments, and flavorings.
  • As used herein, the term “rate controlling polymer material” comprises hydrophilic polymers, hydrophobic polymers and mixtures of hydrophilic and/or hydrophobic polymers that are capable of controlling or retarding the release of the drug compound such as a GnRH related compound from a solid oral dosage form of the present invention. Suitable rate controlling polymer materials include those selected from the group consisting of hydroxyalkyl cellulose such as hydroxypropyl cellulose and hydroxypropyl methyl cellulose; poly(ethylene) oxide; alkyl cellulose such as ethyl cellulose and methyl cellulose; carboxymethyl cellulose, hydrophilic cellulose derivatives; polyethylene glycol; polyvinylpyrrolidone; cellulose acetate; cellulose acetate butyrate; cellulose acetate phthalate; cellulose acetate trimellitate; polyvinyl acetate phthalate; hydroxypropylmethyl cellulose phthalate; hydroxypropylmethyl cellulose acetate succinate; polyvinyl acetaldiethylamino acetate; poly(alkylmethacrylate) and poly (vinyl acetate). Other suitable hydrophobic polymers include polymers and/or copolymers derived from acrylic or methacrylic acid and their respective esters, zein, waxes, shellac and hydrogenated vegetable oils. In one embodiment, the rate-controlling polymer comprises a polymer derived from acrylic or methacrylic acid and their respective esters or copolymers derived from acrylic or methacrylic acid and their respective esters. In another embodiment, the rate-controlling polymer comprises hydroxypropylmethylcellulose (HPMC).
  • In some embodiments, at least one rate controlling polymer material is selected from the group consisting of poly acrylic acid, poly acrylate, poly methacrylic acid and poly methacrylate polymers such as those sold under the Eudragit® trade name (Rohm GmbH, Darmstadt, Germany), for example, Eudragit® L, Eudragit® S, Eudragit® RL, Eudragit® RS coating materials and mixtures thereof. Some of these polymers can be used as delayed release polymers to control the site where the drug is released. In some embodiments, the rate controlling polymer include polymethacrylate polymers such as those sold under the Eudragit® trade name (Rohm GmbH, Darmstadt, Germany), for example, Eudragit® L, Eudragit® S, Eudragit® RL, Eudragit® RS coating materials and mixtures thereof.
  • In some embodiments, the present invention may further comprise diluents. Any suitable diluent may be used in the present invention. Exemplary diluents include, but are not limited to, pharmaceutically acceptable inert fillers such as microcrystalline cellulose, lactose, dibasic calcium phosphate, saccharides, and/or mixtures thereof. Examples of diluents include microcrystalline cellulose such as those sold under the Avicel trademark (FMC Corp., Philadelphia, Pa.), for example, Avicel™ pH101, Avicel™ pH102 and Avicel™ pH112; lactose such as lactose monohydrate, lactose anhydrous and Pharmatose DCL21; dibasic calcium phosphate such as Emeompress® (JRS Pharma, Patterson, N.Y.); mannitol; starch; sorbitol; sucrose; and glucose. In some embodiments, the inert filler comprises microcrystalline cellulose. In one embodiment, the inert filler comprises a lactose selected from the group consisting of lactose monohydrate and lactose anhydrous. In another embodiment, the inert filler comprises a saccharide selected from the group consisting of mannitol, starch, sorbitol, sucrose, and glucose. In one embodiment, the saccharide is sorbitol.
  • In some embodiments, the composition of the present invention may further comprise lubricants. Any suitable lubricant may be used in the present invention. In some embodiments, the lubricant comprises agents that act on the flowability of the powder to be compressed. Exemplary lubricants include, but are not limited to, colloidal silicon dioxide such as Aerosil™ 200, talc, stearic acid, magnesium stearate, and calcium stearate. In some embodiments, the lubricant is stearic acid.
  • In some embodiments, the composition of the present invention may further comprise disintegrants. Any suitable disintegrant may be used in the present invention. Exemplary disintegrants include, but are not limited to, lightly cross-linked polyvinyl pyrrolidone, corn starch, potato starch, maize starch and modified starches, croscarmellose sodium, cross-povidone, sodium starch glycolate and combinations and mixtures thereof. In some embodiments, the disintegrant is chosen from crospovidone and polyvinylpyrrolidone.
  • In some embodiments, the composition described above may further comprise an enhancer. The enhancer can be any suitable enhancer that is known to one of ordinary skill in the art. Exemplary enhancers include, but are not limited to, a medium chain fatty acid salt, ester, ether or a derivative of a medium chain fatty acid which has a carbon chain length of from 4 to 20 carbon atoms. In some embodiments, the enhancer is solid at room temperature. In some embodiments, the enhancer is medium chain fatty acid salt, ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms. In some embodiments, the carbon chain length is from 8 to 14. In some embodiments, the enhancers are S-Cyclodextrins, vitamin E TPGS, gallic acid esters, crospovidones, sorbitan esters, poloxamers, or olyoxyethylene glycolated natural or hydrogenated castor oil (Cremophor®, BASF). In some embodiments, the enhancers are medium chain glycerides or a mixture of medium chain glycerides. Exemplary enhancers are further described in U.S. Patent Publication No. 2003/0091623, and U.S. Pat. No. 6,372,728 which are incorporated by reference in their entireties.
  • IV. Application of Co-Solvent System During the Preparation of a GnRH Related Compound
  • The GnRH related compound described herein may be prepared in the presence of a co-solvent system in a manner that the gelation of the GnRH compound is reduced. In some embodiments, the final step of the preparation of the GnRH related compound is conducted in the presence of a co-solvent system. In some embodiments, the final step is drying, using known techniques, such as lyophilization, tray drying, spray drying, fluid bed drying, or other similar techniques known to one skilled in the art.
  • In some embodiments, the co-solvent system comprises water and at least one water-miscible solvent. The water miscible solvent can be chosen from, but are not limited to, linear or branched C1-6 alcohol, tetrahydrofuran, acetone, ethyl methyl ketone, methyl isobutyl ketone, methyl isopropyl ketone, cyclohexanone, diethyl ketone, pentan-3-one, cyclohexane, acetonitrile, N,N-dimethylformamide, N,N-dimethylacetamide, dioxane, alcohol, ethylene glycol, diglyme, monoglyme, ethylene glycol monomethyl ether, diethylene glycol, triethylene glycol, polyethylene glycol and a mixture thereof. In certain embodiments, at least one water miscible solvent is a linear or branched C1-6 alcohol. Exemplary suitable alcohols include, but are not limited to, methanol, ethanol, propanol, iso-propanol, butanol, sec-butanol, iso-butanol, tert-butanol, 1-pentanol, 2-pentanol, and hexanol. In some embodiments, at least one water miscible solvent is selected from the group consisting of methanol, ethanol, n-propanol, iso-propanol and tert-butanol. In some embodiments, at least one water miscible solvent is selected from the group consisting of methanol, ethanol, iso-propanol, tert-butanol, acetoniltrile and methylene chloride. In some embodiments, the present invention may be carried out with two or more water miscible solvents. In some embodiments, the weight ratio of the water miscible solvent to water is in the range of about 1/1000 to about 99/1. In some embodiments, the weight ratio is about 3/97 to about 59/41.
  • In some embodiments, the process comprises further adding acid during the preparation of the GnRH related compound. Acids used in the present invention include, but are not limited to acetic acid, sulfuric acid, hydrochloride acid, trifluoracetate, citrate acid, tartaric acid, ascorbic acid, and boric acid, etc. Generally, the concentration of the acid is sufficient to prepare a salt of the GnRH related compound. In some embodiments, the concentration of the acid depends on the molecular weight of the GnRH related compound and the acid used herein. In some embodiments, the concentration is in the range of about 0.5% to about 20% of the solution.
  • V. Methods of Preparing the Compositions
  • According to some aspects of the present invention, methods for preparation of a composition of a peptide, polypeptide, protein, an analog or derivative thereof, are provided. In some embodiments, the method comprises mixing the peptide, polypeptide, protein, an analog or derivative thereof with a sufficient amount of at least one stabilizing agents to improve the stability of peptide, polypeptide, protein, an analog and/or derivative thereof. In some embodiments, at least one stabilizing agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent. As used herein, the term “mixing” means contacting, combining, reacting, and/or coating a peptide, polypeptide, protein, an analog and/or derivative thereof, with one or more stabilizing agents. In one embodiment, “mixing” means combining a peptide, polypeptide, protein, an analog or derivative thereof with a stabilizing agent in a composition. In one embodiment, the GnRH related compound is prepared in the presence of a co-solvent system described herein. In some embodiment, the concentration of the stabilizing agent in the composition is equal to or above the critical micelle concentration of the stabilizing agent.
  • VI. Pharmaceutical Compositions and Administration
  • In one embodiment, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of one or more peptide, polypeptide, protein, an analog or derivative thereof, and a sufficient amount of at least one stabilizing agents to improve the stability of the peptide, polypeptide, protein, an analog or derivative thereof, wherein at least one stabilizing agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent. In another embodiment, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier. The term “pharmaceutically acceptable carrier” as used herein refers to any substance, not itself a therapeutic agent, used as a vehicle for delivery of a therapeutic agent to a subject.
  • The compositions of the present invention may be suitable for formulation for oral, parenteral, inhalation spray, topical, rectal, nasal, sublingual, buccal, vaginal or implanted reservoir administration, etc. In one embodiment, the compositions are administered orally, topically, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • In some embodiments, the composition used in the present invention is in an oral dosage form. In some embodiments, the oral dosage form is chosen from tablets, capsules, granules, powders, capsules filled with granules or powders, capsules filled with liquids or semi-solids, sachets filled with granules or powders, liquid, emulsions, microemulsions, and any composition that capable of forming emulsions. In one embodiment, the oral dosage form may be a tablet, a multiparticulate, or a capsule. In one embodiment, the medium chain fatty acid and derivatives thereof described herein may also function as an enhancer.
  • In some embodiments, the oral dosage form is a delayed release dosage form which minimizes the release of the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agents and/or the enhancer in the stomach, and hence the dilution of the local stabilizing agent or the concentration of the enhancer therein, and releases the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent or the enhancer in the intestine. In some embodiments, the oral dosage form is a delayed release rapid onset dosage form. Such a dosage form minimizes the release of peptide, polypeptide, protein, an analog or derivative thereof and stabilizing agent or enhancer in the stomach, and hence the dilution of the local stabilizing agent or enhancer concentration therein, but releases the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent or the enhancer rapidly once the appropriate site in the intestine has been reached, maximizing the delivery of the poorly permeable or soluble peptide, polypeptide, protein, an analog or derivative thereof by maximizing the local concentration of the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent at the site of absorption.
  • In the case of any of the embodiments described herein, a controlled release coating may be applied to the final dosage form (capsule, tablet, multilayer tablet etc.). In one embodiment, the controlled release coating may comprise a rate controlling polymer material described herein. The dissolution characteristics of such a coating material may be pH dependent or independent of pH.
  • In some embodiments, the composition of the present invention may have an enteric coating thereon. In one embodiment, the enteric coating comprises a polymer selected from the group consisting of poly(acrylic acid), polyacrylate, poly(methacrylic acid), polymethacrylate, and mixtures thereof. In some embodiments, the enteric coated composition may be in the form of a tablet or capsule.
  • The term “tablet” as used herein includes, but is not limited to, immediate release (IR) tablets, sustained release (SR) tablets, matrix tablets, multilayer tablets, multilayer matrix tablets, extended release tablets, delayed release tablets and pulsed release tablets any or all of which may optionally be coated with one or more coating materials, including polymer coating materials, such as enteric coatings, rate-controlling coatings, semi-permeable coatings and the like. The term “tablet” also includes osmotic delivery systems in which a drug compound is combined with an osmagent (and optionally other excipients) and coated with a semi-permeable membrane, the semi-permeable membrane defining an orifice through which the drug compound may be released. Tablet solid oral dosage forms may be useful in the practice of the invention include those selected from the group consisting of IR tablets, SR tablets, coated IR tablets, matrix tablets, coated matrix tablets, multilayer tablets, coated multilayer tablets, multilayer matrix tablets and coated multilayer matrix tablets. In some embodiments, a tablet dosage form is an enteric-coated tablet dosage form. In some embodiments, a tablet dosage form is an enteric-coated rapid onset tablet dosage form.
  • Capsule solid oral dosage forms may be useful in the practice of the present invention include those selected from the group consisting of instant release capsules, sustained release capsules, coated instant release capsules, and coated sustained release capsules including delayed release capsules. Capsules may be filled with powders, granules, multi particulates, tablets, semi-solids, or liquids. In some embodiments, a capsule dosage form is an enteric-coated capsule dosage form. In some embodiments, a capsule dosage form is an enteric-coated rapid onset capsule dosage form. Capsules may be made of hard gelatin, soft gelatin, starch, cellulose polymers, or other materials as known to the art.
  • The term “multiparticulate” as used herein means a plurality of discrete particles, pellets, mini-tablets and mixtures or combinations thereof. If the oral form is a multiparticulate capsule, such hard or soft gelatin capsules can suitably be used to contain the multiparticulate. In some embodiments, a sachet may suitably be used to contain the multiparticulate. In some embodiments, the multiparticulate may be coated with a layer containing rate controlling polymer material. In some embodiments, a multiparticulate oral dosage form according to the invention may comprise a blend of two or more populations of particles, pellets, or mini-tablets having different in vitro and/or in vivo release characteristics. For example, a multiparticulate oral dosage form may comprise a blend of an instant release component and a delayed release component contained in a suitable capsule.
  • In some embodiments, the multiparticulate and one or more auxiliary excipient materials can be compressed into tablet form such as a multilayer tablet. In some embodiments, a multilayer tablet may comprise two layers containing the same or different levels of the same active ingredient having the same or different release characteristics. In some embodiments, a multilayer tablet may contain different active ingredient in each layer. Such a tablet, either single layered or multilayered, can optionally be coated with a controlled release polymer so as to provide additional controlled release properties. In some embodiments, multiparticulate dosage form comprises a capsule containing delayed release rapid onset minitablets. In some embodiments, a multiparticulate dosage form comprises a delayed release capsule comprising instant release minitablets. In some embodiments, a multiparticulate dosage form comprises a capsule comprising delayed release granules. In some embodiments, a multiparticulate dosage form comprises a delayed release capsule comprising instant release granules.
  • The term “emulsion” used herein means a suspension or dispersion of one liquid within a second immiscible liquid. In some embodiments, the emulsion is an oil-in-water or water-in-oil-in-water emulsion.
  • The term, “microemulsion” used herein means a solution in which the hydrophobic (oil-like) phase and the hydrophilic (water-like) phase and a surface active agent form micelle structures. Such dispersions are clear and stable over time.
  • In addition, “emulsion” or “microemulsion”, used herein includes a hydrophilic or a hydrophobic liquid which, on dilution with a hydrophobic or a hydrophilic liquid respectively, form an emulsion or a microemulsion. In some embodiments, “emulsion”, or ‘microemulsion’, used herein may include solid or semi-solid materials which may be liquid at higher temperatures. For example, the material may be solid at room temperature. At about body temperature (about 37° C.), the material may be liquid.
  • Alternatively, pharmaceutically acceptable compositions of this invention may be in the form of a suppository for rectal administration. The suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
  • Pharmaceutically acceptable compositions of the present invention may be in the form of a topical solution, ointment, or cream in which the active component is suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Where the topical formulation is in the form of an ointment or cream, suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2 octyldodecanol, benzyl alcohol and water.
  • When the pharmaceutically acceptable composition is an ophthalmic formulation, it may be a micronized suspension in isotonic, pH adjusted sterile aqueous solution, or as a solution in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in the form of an ointment.
  • The pharmaceutically acceptable compositions of this invention may also be administered by nasal, aerosol or by inhalation administration routes. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, gender, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • VII. Methods of Treatment and Use
  • Another aspect of the present invention provides a method of treatment of a medical condition treatable by a peptide, polypeptide, protein, an analog or derivative thereof comprising administering to a patient suffering from said condition a pharmaceutical composition as described in the present application. As used herein, the medical condition includes, but is not limited to, diabetes mellitus, Alzheimer, autoimmune diseases, colon cancer, sex hormone dependent disease such as benign prostate hyperplasia, prostate cancer, estrogen-dependent breast cancer, endometrial cancer, ovarian cancer, endometriosis and precocious puberty, and contraception in a human or animal subject
  • Yet, another embodiment provides use of pharmaceutical composition as described in the present application in the manufacture of a medicament for the treatment of a medical condition treatable by said peptide, polypeptide, protein, analogs and/or derivatives thereof.
  • It is understood that the combinations of all embodiments described herein are also envisaged in the present invention.
  • The present invention will now be described in more detail with reference to the following examples. However, these examples are given for the purpose of illustration and are not to be construed as limiting the scope of the invention.
  • EXAMPLES 1. Application of Co-Solvent During the Preparation of Acyline (1) Preparation of Acyline Batches
  • The six different acyline batches are described in FIG. 1, which summarizes the variations in the lyophilization procedure, which is the last step of the preparation of acyline. For lots XF 173/315-125, XF 185/165-133 and XF 173/315-151A, a co-solvent system is used in the lyophilization step. Water is used as a solvent for other batches. The batches and the associated lyophilization solvents are also illustrated in FIG. 1. The acyline may be prepared according to methods known to one of skill in the art, for example U.S. Pat. No. 6,747,125, or processes described in the present application.
  • Example 1
  • The study of gelation for different acyline batches in propylene glycol is carried out and the result is summarized in FIG. 2. The 9% and 16.7% of acyline sampls are prepared by using the acyline batches listed in FIG. 1. The 9% acyline sample of XF 173/315-125, XF 185/165-133 and XF 173/315-151A are prepared by using acyline prepared via use of a co-solvent during the lyophilization step. The acyline batches of XF 173/315-125, XF 185/165-133 and XF 173/315-151A appear clear and non-viscous after 2 hours. Other batches which are lyophilized by using water as a solvent did not appear as clear and non-viscous solutions due to the presence of gelled acyline.
  • Example 2
  • The gelation of different acyline drug batches in water is investigated and the results are summarized in FIG. 3. The 0.5% and 1% acyline samples are prepared by using the acyline batches listed in FIG. 1. The 0.5% and 1% of acyline samples of XF 173/315-125, XF 185/165-133 and XF 173/315-151A which are prepared by using co-solvent during the lyophilization step appear clear and non viscous upon dissolving in water.
  • Example 3
  • The tendency of gelation of different acyline batches in standardized microemulsion (SM) is investigated. The result is summarized in FIG. 4( a). The formulation of the standard micro emulsion is shown in FIG. 4( b). The 5 mg and 10 mg formulations of acyline in a microemulsion are prepared by using the acyline lots synthesized in FIG. 1. 5 mg and 10 mg dose of acyline batches of XF 173/315-151A which are prepared by using co-solvent during the lyophilization step appeared clear and transparent after 2 hours.
  • 2. Application of Anti-Gelling Agents in the Formulation of Acyline Compositions General Procedures of Comparison Experiments
  • Different concentrations of acyline composition are prepared by adding acyline to pH 6.8 buffer solution either at room temperature or 37° C. All acyline solutions contain 0.6 mg/mL sodium caprate (sodium caprate is referred as C10 in the figures). All samples are centrifuged and filtered prior to analysis. Samples are analyzed by reverse phase HPLC with UV detection.
  • Example 4
  • 10 mg acyline is transferred into 200 mL volumetric flask. A pH 6.8 buffer solution is preheated to 37° C. Then, 100 mL pre-heated buffer solution and 0.6 mg/mL sodium caprate is added to the flask to prepare a 0.1 mg/mL acyline sample. In a similar manner, Tween 80 is added to prepare a 0.1 mg/mL acyline solution with 1% Tween 80. Samples are shaken in a temperature controlled water bath at 37° C. 5 mL samples are taken at 1, 5, 10, 15, 20, 30 and 120 minutes after mixing acyline with the buffer solution. Samples are filtered immediately through 0.45 μm filters and the first 3 mL is discarded. Filtered samples are analyzed, undiluted, by reverse phase HPLC with UV detection. All samples are prepared in duplicate and the concentration was obtained for the analysis from the mean of the duplicates. The result of comparison of 1% and 0% Tween 80 at 0.1 mg/mL acyline sample is graphically recorded in FIG. 5. The results show that sodium caprate alone at a concentration below the CMC is insufficient to reduce the gelation of acyline. The addition of 1% Tween 80 successfully reduces the gelation.
  • Example 5
  • A similar experimental procedure as Example 4 is used to prepare acyline sample for example 5. The result of comparison of 5 mg/mL, 1.0 mg/mL, 0.1 mg/mL, and 0.1% Tween in phosphate buffer containing 0.6 mg/mL of sodium caprate and 0.1 mg/mL acyline is graphically recorded in FIG. 6. The results show that only 1% Tween 80 solution may completely inhibit the gelation of acyline.
  • Example 6
  • A similar experimental procedure as Example 4 is used to prepare acyline sample for example 6. The result of comparison of 1% and 1 mg/mL Tween 80 in 0.1 and 0.01 mg/mL acyline sample is graphically recorded in FIG. 7. The results show that 1% Tween 80 reduces gelation for both 0.1 mg/mL and 0.01 mg/mL acyline sample.
  • Example 7
  • A similar experimental procedure as Example 4 is used to prepare acyline sample for example 7. The result of comparison of 0.01 mg/mL acyline samples having 0.1%, 0.5% and 1% Tween 80 is graphically recorded in FIG. 8. The results show that the acyline sample with 1% Tween 80 significantly reduces the gelation.
  • 3. Impact of Sodium Caprate (C10) on the Tendency of Gelation of Acyline in Water Example 8
  • FIG. 9 graphically demonstrates the impact of different concentrations of sodium caprate (C10) on the gelation of acyline in water. When the concentration of sodium caprate is below 10 mg/mL, the recovery of acyline significantly decreases, which implies an increase of gelation of acyline. The investigators of the present invention believe that the increase of the gelation is due to an increased concentration of ions caused by the addition of sodium caprate. However, when the concentration of sodium caprate reaches and is above the CMC of sodium caprate (˜20 mg/mL), it is observed that there is a sudden and significant increase of the recovery of acyline, which indicates an effective reduction of gelation. The CMC of sodium caprate is known as 100 mM (˜20 mg/mL). (See “kinetic studies of the interaction of fatty acids with phosphatidylcholine vesicles (liposomes), Rogerson et al., Colloids and Surfaces B: Biointerfaces, 48, 24-34 (2006).) When the concentration of sodium caprate reaches about 50 mg/mL, the recovery of acyline is almost 100%, which indicates that the gelation is completely inhibited.
  • 4. Application of Anti-Gelling Agents in Different Micoremulsion formulations of Acyline Samples
  • Example 9
  • FIG. 11 graphically demonstrates the relative bioavailability of various formulations of acyline in dogs. The relative bioavailability is measured by comparing the absolute bioavailability of various formulations of acyline with the absolute bioavailability of a standard formulation of acyline, which is a formulation without any anti-gelling agent. The formulation of microemulsion 1 (M1/55% Capmul MCM), microemulsion 2 (M2/45% Capmul PG-8), and microemulsion 3 (M3/55% Capmul MCM C10) are illustrated in FIG. 10( a) through (c). The formulation of the standard solution is 5 mg acyline, 550 mg sodium caprate and 5 mL purified water. “C10” in FIGS. 10 and 11 represents sodium caprate. “SLS” in FIGS. 10 and 11 represents sodium lauryl sulphate. The comparison results in FIG. 11 show that all formulations with anti-gelling agents such as sodium caprate and sodium lauryl sulphate, increase the bioavailability of acyline from 7.8 fold to 32.5 fold.
  • Example 10
  • FIG. 12 graphically demonstrates the relative bioavailability of (1) enteric tablets of 10 mg acyline versus unenhanced 5 mg acyline solution sample and (2) enteric tablets of 10 mg acyline versus 5 mg acyline standard sample. The formulation of acyline standard sample is 5 mg acyline, 550 mg sodium caprate and 5 mL purified water. The acyline in the tablets and the 5 mg acyline sample is prepared by using a lyophilization step with water as the solvent. The tablets contained the same amount of sodium caprate as the 5 mg acyline sample. It is observed that, the concentration of sodium caprate, 110 mg/mL, is sufficient to reduce or even inhibit gelation in the solution and enhance the bioavailability of acyline solution. FIG. 12 demonstrates that the bioavailability of enhanced tablets is significantly improved compared to unenhanced tablet.
  • 5. Application of Sodium Caprate and Capric Acid in Insulin Samples Example 11
  • A study evaluating the effect of sodium caprate (C10) and capric acid on the stability of bovine insulin is carried out. In this study, bovine Insulin with a potency of 29 U/mg is used to prepare a 10 U/mL solution (344.83 mg/L of solution) in a phosphate buffer pH 8. Using 200 mL vessels, appropriate amounts of C10 is added to 100 mL aliquots of the insulin solution. Two bottles are used a controls, containing only the insulin solution. An addition of 3 g of C10 is added to bottles 3 and 4, 6 g of C10 to bottles 5 and 6, 3 g of Capric Acid to bottles 7 and 8, and 6 g of Capric Acid to bottles 9 and 10. The bottles are gently swirled and 6 mL aliquots are removed from each bottle for the T=ZERO sample. The samples are filtered (Millex-HV 0.45 μm), and the first 5 ml of filtrate discarded. The appropriate test solutions are stirred and additional samples are collected from all test solutions at 6, 24 hours etc. The amount of bovine insulin in solution after filtration is determined against standards (70 mg in 200 ml of 0.01 N HCl).
  • The % recovery of intact insulin after the insulin solution stands for a period of time is recorded in Tables 2 and 3 and graphically illustrated in FIGS. 13( a) and (b). It is observed that insulin solutions with sodium caprate are significantly more stable than the insulin samples without sodium caprate or the solutions with capric acid. For insulin samples that contain sodium caprate, at least 90% of insulin is recovered after the solutions stands at 37° C. for at least about 24 hours.
  • TABLE 1
    Formulation of insulin solutions for Example 11
    10 U/mL
    Bovine
    Insulin in
    ‘blank’
    simulated 10 U/mL Bovine Insulin in ‘blank’ 10 U/mL Bovine Insulin in ‘blank’
    intestinal simulated intestinal buffer pH 8 simulated intestinal buffer pH 8
    buffer pH 8 Plus Sodium Caprate Plus Capric Acid
    1 3 5 7 9
    Sample Control 2 Control 4 Control 6 Control 8 Control 10
    Sodium 30 mg/ml 30 mg/ml 60 mg/ml 60 mg/ml
    Caprate
    Capric 30 mg/ml 30 mg/ml 60 mg/ml 60 mg/ml
    Acid
    Stirring No Yes No Yes No Yes No Yes No Yes
  • TABLE 2
    Bovine insulin fibrillation results with or without C10
    % Recovery % Recovery % Recovery % Recovery % Recovery % Recovery
    T = T = T = T = T = T =
    Sample 0 6 24 29 48 72
    1. Insulin Standing 100.24 100.00 100.25 100.28 100.29 100.29
    2. Insulin Stirring 100.11 100.08 99.42 1.50 0.29 0.22
    3. 30 mg/ml C10 98.15 97.85 97.88 97.39 97.15 96.54
    Standing
    4. 30 mg/ml C10 98.06 97.66 97.74 97.44 97.58 97.05
    Stirring
    5. 60 mg/ml C10 94.52 94.24 93.99 93.61 93.24 92.12
    Standing
    6. 60 mg/ml C10 94.79 94.52 94.41 93.98 93.29 92.79
    Stirring
  • TABLE 3
    Bovine insulin fibrillation results with or without Capric acid
    Bovine Insulin Fibrillation Expt. Results Summary
    % Recovery % Recovery % Recovery % Recovery % Recovery % Recovery
    T = T = T = T = T = T =
    Sample 0 6 24 29 48 72
    1. Insulin Standing 100.24 100.00 100.25 100.28 100.29 100.29
    2. Insulin Stirring 100.11 100.08 99.42 1.50 0.29 0.22
    7. 30 mg/ml CA 93.73 68.88 15.32 3.26 1.10 0.44
    Standing
    8. 30 mg/ml CA 95.06 0.27 0.20 0.24 0.23 0.16
    Stirring
    9. 60 mg/ml CA 91.96 67.73 19.75 2.59 1.43 2.24
    Standing
    10. 60 mg/ml CA 91.38 0.27 0.19 0.09 0.13 0.10
    Stirring
    Note:
    Recovery values based on the theoretical amount spiked = 0.344591 mg/ml
  • The foregoing is illustrative of the present invention and is not to be construed as limiting thereof. Although a few exemplary embodiments of this invention have been described, those skilled in the art will readily appreciate that many modifications are possible in the exemplary embodiments without materially departing from the novel teachings and advantages of this invention. Accordingly, all such modifications are intended to be included within the scope of this invention as defined in the claims. Therefore, it is to be understood that the foregoing is illustrative of the present invention and is not to be construed as limited to the specific embodiments disclosed, and that modifications to the disclosed embodiments, as well as other embodiments, are intended to be included within the scope of the appended claims. The invention is defined by the following claims, with equivalents of the claims to be included therein.

Claims (28)

1. A composition comprising a therapeutically effective amount of at least one peptide, polypeptide, protein, an analog or derivative thereof and a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof,
wherein at least one stabilizing agent is a salt, an ester, an ether, or a derivative of a medium chain fatty acid and having a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent.
2. The composition of claim 1, wherein at least 90% of the peptide, polypeptide, protein, analog or derivative thereof is retained after the composition stands at 37° C. for at least about 24 hours.
3. The composition of claim 1, wherein at least one peptide, polypeptide, protein, analog or derivative thereof is insulin, or an analog or derivative thereof.
4. The composition of claim 1, wherein at least one peptide, polypeptide, protein, analogs or derivative thereof is glucagon-Like Peptide (GLP), or an analog or derivative thereof.
5. The composition of claim 4, wherein at least one GLPs is selected from the group consisting of GLP-1, GLP-1 analogs and derivatives thereof, GLP-2, GLP-2 analogs and derivatives thereof, exendin-4, analogs and derivatives thereof.
6. The composition of claim 1, wherein at least one peptide, polypeptide, protein, an analog or derivative thereof is selected from the group consisting of GnRH agonists and antagonists, somatostatin, ACTH, corticotropin-releasing factor, angiotensin, calcitonin, gastric inhibitory peptide, growth hormone-releasing factor, pituitary adenylate, exendin, exendin-3, cyclase activating peptide, secretin, enterogastrin, somatostatin, somatotropin, somatomedin, parathyroid hormone, thrombopoietin, erythropoietin, hypothalamic releasing factors, prolactin, thyroid stimulating hormones, endorphins, enkephalins, vasopressin, oxytocin, opioids and analogues thereof, superoxide dismutase, interferon, asparaginase, arginase, arginine deaminase, adenosine deaminase, ribonuclease, FVII, FXIII, a mixture of FVII and FXIII, IL-20, IL-21, IL-28a, IL-29, IL-31, and analogs and derivatives thereof.
7. The composition of claim 1, which is solid at room temperature.
8. The composition of claim 1, which is in a solid oral dosage form.
9. The composition of claim 1, wherein the stabilizing agent is a salt of a medium chain fatty acid and has a carbon chain length of from about 8 to 14 carbon atoms.
10. The composition of claim 1, wherein the stabilizing agent is selected from the group consisting of sodium caprylate, sodium caprate and sodium laurate.
11. The composition of claim 1, wherein the concentration of the stabilizing agent in the composition is equal to or above the critical micelle concentration of the stabilizing agent in the composition.
12. The composition of claim 1, wherein the dissolution rate of the stabilizing agent and the peptide, polypeptide, protein, analogs or derivatives thereof in the composition are substantially the same.
13. The composition of claim 1, further comprising one or more excipients selected from the group consisting of rate-controlling polymeric materials, diluents, lubricants, disintegrants, plasticizers, anti-tack agents, opacifying agents, pigments, and flavorings.
14. The composition of claim 13, wherein at least one rate-controlling polymer is a polymer derived from acrylic or methacrylic acid, esters or copolymers derived from acrylic or methacrylic acid.
15. The composition of claim 1, further comprising an enteric coating.
16. The composition of claim 15, wherein the enteric coating comprises at least one polymer selected from the group consisting of poly(acrylic acid), polyacrylate, poly(methacrylic acid), polymethacrylate, and mixtures thereof.
17. The composition of claim 15, wherein the enteric coated composition is a tablet or capsule.
18. The composition of claim 1, wherein the composition is in a form selected from the group consisting of a multiparticulate form, a sustained-release form and an instant release form.
19. The composition of claim 1, further comprising at least one diluent which is an inert filler chosen from microcrystalline cellulose, lactose, dibasic calcium phosphate and saccharides.
20. The composition of claim 19, wherein the inert filler is at least one lactose which is lactose monohydrate or lactose anhydrous.
21. The composition of claim 20, wherein the inert filler is at least one saccharide selected from the group consisting of mannitol, starch, sorbitol, sucrose, and glucose.
22. The composition of claim 1, further comprising at least one lubricant selected from the group consisting of colloidal silicon dioxide, talc, magnesium stearate, calcium stearate, and stearic acid.
23. The composition of claim 1, further compriseing at least one disintegrant selected from the group consisting of lightly crosslinked polyvinylpyrrolidone, corn starch, potato starch, maize starch and modified starches, croscarmellose sodium, crospovidone, and sodium starch glycolate.
24. A method for preparation of a composition of a peptide, polypeptide, protein, an analog or derivative thereof, wherein the process comprises mixing the peptide, polypeptide, protein analogs or derivatives thereof with a sufficient amount of at least one stabilizing agents to improve the stability of peptide, polypeptide, protein, an analog or derivative thereof, and the agent is a salt, an ester, an ether, or a derivative of a medium chain fatty acid and having a carbon chain length of from about 4 to about 20 carbon atoms, or is a surface active agent.
25. The methods of claim 24, wherein at least 90% of the peptide, polypeptide, protein, analog or derivative thereof is retained after the composition stands at 37° C. for at least about 24 hours.
26. The method of claim 24, wherein the stabilizing agent is a medium chain fatty acid salt.
27. The method of claim 24, wherein the stabilizing agent is a medium chain fatty acid salt having a carbon chain length of from about 8 to about 14 carbon atoms.
28. The method of claim 24, wherein the concentration of the stabilizing agent in the composition is equal or above the critical micelle concentration of the stabilizing agent in the composition.
US12/436,990 2008-05-07 2009-05-07 Compositions of peptides and processes of preparation thereof Abandoned US20090280169A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/436,990 US20090280169A1 (en) 2008-05-07 2009-05-07 Compositions of peptides and processes of preparation thereof
TW098115182A TW200950801A (en) 2008-05-07 2009-05-07 Compositions of peptides and processes of preparation thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5103808P 2008-05-07 2008-05-07
US12/436,990 US20090280169A1 (en) 2008-05-07 2009-05-07 Compositions of peptides and processes of preparation thereof

Publications (1)

Publication Number Publication Date
US20090280169A1 true US20090280169A1 (en) 2009-11-12

Family

ID=40801968

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/436,990 Abandoned US20090280169A1 (en) 2008-05-07 2009-05-07 Compositions of peptides and processes of preparation thereof
US12/437,012 Expired - Fee Related US8999383B2 (en) 2008-05-07 2009-05-07 Compositions of GnRH related compounds and processes of preparation
US14/636,756 Abandoned US20150238561A1 (en) 2008-05-07 2015-03-03 Compositions of GnRH related compounds and processes of preparation

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/437,012 Expired - Fee Related US8999383B2 (en) 2008-05-07 2009-05-07 Compositions of GnRH related compounds and processes of preparation
US14/636,756 Abandoned US20150238561A1 (en) 2008-05-07 2015-03-03 Compositions of GnRH related compounds and processes of preparation

Country Status (14)

Country Link
US (3) US20090280169A1 (en)
EP (2) EP2288369B1 (en)
JP (4) JP2011519928A (en)
KR (2) KR20110007614A (en)
CN (3) CN102046192B (en)
AU (2) AU2009244797B2 (en)
BR (1) BRPI0912384A2 (en)
CA (2) CA2723558A1 (en)
HK (1) HK1157185A1 (en)
IL (1) IL209136A (en)
MX (1) MX2010012201A (en)
RU (1) RU2517135C2 (en)
TW (2) TW200950799A (en)
WO (2) WO2009137078A1 (en)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070148228A1 (en) * 1999-02-22 2007-06-28 Merrion Research I Limited Solid oral dosage form containing an enhancer
US20070196464A1 (en) * 1999-02-22 2007-08-23 Merrion Research I Limited Solid oral dosage form containing an enhancer
US20070219131A1 (en) * 2004-04-15 2007-09-20 Ben-Sasson Shmuel A Compositions capable of facilitating penetration across a biological barrier
US20080159984A1 (en) * 2004-04-15 2008-07-03 Ben-Sasson Shmuel A Compositions Capable of Facilitating Penetration Across a Biological Barrier
US20100209499A1 (en) * 1999-02-22 2010-08-19 Cumming Kenneth I Solid Oral Dosage Form Containing an Enhancer
US8329198B2 (en) 2008-09-17 2012-12-11 Chiasma Inc. Pharmaceutical compositions and related methods of delivery
US8802114B2 (en) 2011-01-07 2014-08-12 Merrion Research Iii Limited Pharmaceutical compositions of iron for oral administration
US8883203B2 (en) 2006-04-07 2014-11-11 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US8999383B2 (en) 2008-05-07 2015-04-07 Merrion Research Iii Limited Compositions of GnRH related compounds and processes of preparation
US9089484B2 (en) 2010-03-26 2015-07-28 Merrion Research Iii Limited Pharmaceutical compositions of selective factor Xa inhibitors for oral administration
US9375478B1 (en) 2015-01-30 2016-06-28 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9670261B2 (en) 2012-12-21 2017-06-06 Sanofi Functionalized exendin-4 derivatives
US9687526B2 (en) 2015-01-30 2017-06-27 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9694053B2 (en) 2013-12-13 2017-07-04 Sanofi Dual GLP-1/glucagon receptor agonists
US9744209B2 (en) 2015-01-30 2017-08-29 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9751926B2 (en) 2013-12-13 2017-09-05 Sanofi Dual GLP-1/GIP receptor agonists
US9750785B2 (en) 2015-01-30 2017-09-05 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9750788B2 (en) 2013-12-13 2017-09-05 Sanofi Non-acylated exendin-4 peptide analogues
US9758561B2 (en) 2014-04-07 2017-09-12 Sanofi Dual GLP-1/glucagon receptor agonists derived from exendin-4
US9771406B2 (en) 2014-04-07 2017-09-26 Sanofi Peptidic dual GLP-1/glucagon receptor agonists derived from exendin-4
US9775904B2 (en) 2014-04-07 2017-10-03 Sanofi Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
US9789165B2 (en) 2013-12-13 2017-10-17 Sanofi Exendin-4 peptide analogues as dual GLP-1/GIP receptor agonists
US9919026B2 (en) 2015-01-30 2018-03-20 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
US9937223B2 (en) 2015-01-30 2018-04-10 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9982029B2 (en) 2015-07-10 2018-05-29 Sanofi Exendin-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
US10265384B2 (en) 2015-01-29 2019-04-23 Novo Nordisk A/S Tablets comprising GLP-1 agonist and enteric coating
US10758592B2 (en) 2012-10-09 2020-09-01 Sanofi Exendin-4 derivatives as dual GLP1/glucagon agonists
US10806797B2 (en) 2015-06-05 2020-10-20 Sanofi Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate
US11338011B2 (en) 2015-02-03 2022-05-24 Amryt Endo, Inc. Method of treating diseases
US11890316B2 (en) 2020-12-28 2024-02-06 Amryt Endo, Inc. Oral octreotide therapy and contraceptive methods

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2011341637A1 (en) * 2010-12-15 2013-06-20 Merrion Research Iii Limited Pharmaceutical compositions of selective factor Xa inhibitors for oral administration
MX364010B (en) * 2012-06-01 2019-04-11 Ferring Bv Manufacture of degarelix.
CN104072605B (en) * 2013-03-27 2017-03-29 深圳翰宇药业股份有限公司 A kind of preparation method for degree Shandong peptide
CN104072603B (en) * 2013-03-27 2017-09-05 深圳翰宇药业股份有限公司 It is a kind of to synthesize for the method for degree Shandong peptide
US10105442B2 (en) * 2014-05-07 2018-10-23 Novo Nordisk A/S Treatment of diabetes type 1 using GLP-1 and anti-IL-21
CN105636937B (en) 2015-01-27 2018-07-17 深圳华大生命科学研究院 Stabilizer for preserving biological sample
CN104998251A (en) * 2015-07-08 2015-10-28 哈尔滨吉象隆生物技术有限公司 Intestinal absorption promoting liraglutide salt for preparing oral enteric-coated preparations
CN105749245A (en) * 2016-03-02 2016-07-13 张光泉 Anti-cancer drug degarelix acetate injection and preparation method thereof
KR101786914B1 (en) * 2017-01-31 2017-11-15 주식회사 삼양사 Adhesive elastic band for skin-lifting or enhancement of skin elasticity
WO2018230751A1 (en) * 2017-06-14 2018-12-20 주식회사 바이오솔루션 Cosmetic composition for wrinkle reduction or anti-inflammation, containing substance p
CA3071660A1 (en) * 2017-08-01 2019-02-07 Fund Sa Adjuvant therapy for use in prostate cancer treatment
CN107412739B (en) * 2017-08-09 2020-02-07 南通大学 New use of IFN-lambda in Zika virus infection
US20190054027A1 (en) * 2017-08-18 2019-02-21 Abbvie Inc. Solid Pharmaceutical Formulations for Treating Endometriosis, Uterine Fibroids, Polycystic Ovary Syndrome or Adenomyosis
SG11202001439QA (en) * 2017-08-18 2020-03-30 Abbvie Inc Solid pharmaceutical formulations for treating endometriosis, uterine fibroids, polycystic ovary syndrome and adenomyosis
WO2019036712A1 (en) * 2017-08-18 2019-02-21 Abbvie Inc. Pharmaceutical formulations for treating endometriosis, uterine fibroids, polycystic ovary syndrome or adenomyosis
CA3073229A1 (en) * 2017-08-18 2019-02-21 Abbvie Inc. Pharmaceutical formulations for treating endometriosis, uterine fibroids, polycystic ovary syndrome or adenomyosis
CN107952063A (en) * 2017-12-27 2018-04-24 北京四环生物制药有限公司 Calcitonin piece and preparation method thereof
AU2018419533A1 (en) 2018-04-19 2020-11-12 Abbvie Inc. Methods of treating heavy menstrual bleeding
CN109400700A (en) * 2018-11-06 2019-03-01 安徽怡发生物科技有限公司 A kind of preparation method of the collagen oligopeptide easily absorbed
CN111265654B (en) * 2018-12-05 2023-05-16 江苏恒瑞医药股份有限公司 Pharmaceutical composition of GLP-1 analogue and preparation method thereof
WO2021180862A1 (en) 2020-03-12 2021-09-16 Synthon B.V. Pharmaceutical compositions comprising elagolix sodium

Citations (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4525339A (en) * 1982-10-15 1985-06-25 Hoffmann-La Roche Inc. Enteric coated oral dosage form
US4590062A (en) * 1984-04-16 1986-05-20 Tech Trade Corp. Dry direct compression compositions for controlled release dosage forms
US4654155A (en) * 1985-03-29 1987-03-31 Reynolds Metals Company Microemulsion lubricant
US4656161A (en) * 1983-08-27 1987-04-07 Basf Aktiengesellschaft Increasing the enteral absorbability of heparin or heparinoids
US4764375A (en) * 1985-09-11 1988-08-16 Kv Pharmaceutical Company Sachet drug delivery system
US4900730A (en) * 1981-01-14 1990-02-13 Toyo Jozo Co., Ltd. Preparation which promotes the absorption of peptides
US4996058A (en) * 1987-09-18 1991-02-26 Ciba-Geigy Corporation Covered retard forms
US5110606A (en) * 1990-11-13 1992-05-05 Affinity Biotech, Inc. Non-aqueous microemulsions for drug delivery
US5190748A (en) * 1988-11-22 1993-03-02 Hoffmann-La Roche Inc. Absorption enhancement of antibiotics
US5221734A (en) * 1987-10-01 1993-06-22 Ciba-Geigy Corporation Process for preparing a polypeptide growth factor for milk
US5229103A (en) * 1992-04-30 1993-07-20 Hydrodent Laboratories, Inc. Antiplaque mouthwash concentrate
US5229130A (en) * 1991-12-20 1993-07-20 Cygnus Therapeutics Systems Vegetable oil-based skin permeation enhancer compositions, and associated methods and systems
US5288497A (en) * 1985-05-01 1994-02-22 The University Of Utah Compositions of oral dissolvable medicaments
WO1994008599A1 (en) * 1992-10-14 1994-04-28 The Regents Of The University Of Colorado Ion-pairing of drugs for improved efficacy and delivery
US5346701A (en) * 1993-02-22 1994-09-13 Theratech, Inc. Transmucosal delivery of macromolecular drugs
US5444041A (en) * 1991-04-19 1995-08-22 Ibah, Inc. Convertible microemulsion formulations
US5506207A (en) * 1994-03-18 1996-04-09 The Salk Institute For Biological Studies GNRH antagonists XIII
US5541155A (en) * 1994-04-22 1996-07-30 Emisphere Technologies, Inc. Acids and acid salts and their use in delivery systems
US5631347A (en) * 1995-06-07 1997-05-20 Eli Lilly And Company Reducing gelation of a fatty acid-acylated protein
US5639469A (en) * 1994-06-15 1997-06-17 Minnesota Mining And Manufacturing Company Transmucosal delivery system
US5650386A (en) * 1995-03-31 1997-07-22 Emisphere Technologies, Inc. Compositions for oral delivery of active agents
US5707648A (en) * 1993-11-17 1998-01-13 Lds Technologies, Inc. Transparent liquid for encapsulated drug delivery
US5714477A (en) * 1993-06-18 1998-02-03 Pharmacia & Upjohn Aktiebolag Pharmaceutical composition containing heparin, heparin fragments or their derivatives in combination with glycerol esters
US5736161A (en) * 1993-07-21 1998-04-07 Lipotec S.A. Pharmaceutical preparation for improving the bioavailability of drugs which are difficult to absorb and a procedure for obtaining it
US5807983A (en) * 1995-12-28 1998-09-15 The Salk Institute For Biological Studies GNRH antagonist betides
US5821222A (en) * 1992-06-11 1998-10-13 Bayer Aktiengesellschaft Cyclic depsipeptides having 18 ring atoms for combating endoparasites
US5821230A (en) * 1997-04-11 1998-10-13 Ferring Bv GnRH antagonist decapeptides
US5863555A (en) * 1995-10-23 1999-01-26 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
US5912009A (en) * 1996-10-30 1999-06-15 Theratech, Inc. Fatty acid esters of glycolic acid and its salts
US6015801A (en) * 1997-07-22 2000-01-18 Merck & Co., Inc. Method for inhibiting bone resorption
US6017944A (en) * 1997-10-28 2000-01-25 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6017559A (en) * 1994-07-15 2000-01-25 Dow Agrosciences Llc Preparation of aqueous emulsions
US6025366A (en) * 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6068850A (en) * 1996-07-03 2000-05-30 Alza Corporation Aqueous formulations of peptides
US6077847A (en) * 1998-04-02 2000-06-20 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6077858A (en) * 1997-06-05 2000-06-20 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6124261A (en) * 1996-07-03 2000-09-26 Alza Corporation Non-aqueous polar aprotic peptide formulations
US6200602B1 (en) * 1995-08-08 2001-03-13 West Pharmaceutical Services Drug Delivery & Clinical Research Centre Limited Composition for enhanced uptake of polar drugs from the colon
US6214798B1 (en) * 1997-04-11 2001-04-10 Ferring Bv GnRH antagonists being modified in positions 5 and 6
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6270804B1 (en) * 1998-04-03 2001-08-07 Biovail Technologies Ltd. Sachet formulations
US6296881B1 (en) * 1998-08-03 2001-10-02 Kenkohyakunijussai Co., Ltd. Bactericide containing iron ions
US20020002140A1 (en) * 2000-01-14 2002-01-03 Holick Michael F. Novel bisphosphonates and uses thereof
US6372728B1 (en) * 1997-10-10 2002-04-16 Astrazeneca Ab Formulation for treatment of osteoporosis
US6379960B1 (en) * 2000-12-06 2002-04-30 Isis Pharmaceuticals, Inc. Antisense modulation of damage-specific DNA binding protein 2, p48 expression
US20030031757A1 (en) * 2001-08-03 2003-02-13 Kraft Food Holdings, Inc. Stable and bioavailable iron fortified beverages
US6524557B1 (en) * 1994-12-22 2003-02-25 Astrazeneca Ab Aerosol formulations of peptides and proteins
US20030091623A1 (en) * 1999-02-22 2003-05-15 Cumming Kenneth Iain Solid oral dosage form containing an enhancer
US20030100509A1 (en) * 2001-11-27 2003-05-29 Werner Sarlikiotis Injectable solution of an LHRH antagonist
US20030114525A1 (en) * 2000-11-21 2003-06-19 Kammer Gary M. Method of treating autoimmune diseases
US20030139378A1 (en) * 2001-12-13 2003-07-24 Daifotis Anastasia G. Liquid bisphosphonate formulations for bone disorders
US20030166508A1 (en) * 2000-06-07 2003-09-04 Junshou Zhang Biologically active oral preparation that can be site-specific released in colon
US20030176397A1 (en) * 2000-04-07 2003-09-18 Lichtenberger Lenard M. Unique compositions of zwitterionic phospholipids and bisphosphonates and use of the compositions as bisphosphate delivery systems with reduced GI toxicity
US20030181421A1 (en) * 2000-06-20 2003-09-25 Horowitz Zebulun D. Method of administering bisphosphonates
US20040087631A1 (en) * 2002-03-04 2004-05-06 Bacopoulos Nicholas G. Methods of treating cancer with HDAC inhibitors
US6747014B2 (en) * 1997-07-01 2004-06-08 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
US6747125B1 (en) * 1991-03-14 2004-06-08 The Salk Institute For Biological Studies Peptide intermediates for making GnRH antagonists
US20040147484A1 (en) * 2001-03-01 2004-07-29 Boyd Maria Aurora P. Compositions for delivering bisphosphonates
US20040157799A1 (en) * 2001-05-02 2004-08-12 Seaman John J Pharmaceutical uses of bisphosphonates
US20050065117A1 (en) * 2003-09-19 2005-03-24 Pfizer Inc Pharmaceutical compositions and methods comprising combinations of 2-alkylidene-19-nor-vitamin D derivatives and a bisphosphonate
US6875843B2 (en) * 2001-01-26 2005-04-05 Children's Mercy Hospital Prevention of diabetes and prolongation of the honeymoon phase of diabetes by administration of GnRH antagonists
US20050080075A1 (en) * 2003-08-25 2005-04-14 Nichols M. James Formulations, conjugates, and combinations of drugs for the treatment of neoplasms
US20050119331A1 (en) * 2003-11-04 2005-06-02 Jackie Butler Pharmaceutical formulations for carrier-mediated transport statins and uses thereof
US20050157799A1 (en) * 2004-01-15 2005-07-21 Arvind Raman System, method, and apparatus for error concealment in coded video signals
US20050163849A1 (en) * 2003-10-31 2005-07-28 Wong Patrick S. Compositions and dosage forms for enhanced absorption of iron
US20050249799A1 (en) * 2004-03-03 2005-11-10 Spherics, Inc. Polymeric drug delivery system for hydrophobic drugs
US20060018874A1 (en) * 2004-07-19 2006-01-26 Balasingam Radhakrishnan Fatty acid formulations for oral delivery of proteins and peptides, and uses thereof
US20060135405A1 (en) * 2004-12-22 2006-06-22 Zentaris Gmbh Process for producing sterile suspensions of slightly soluble basic peptide complexes, sterile suspensions of slightly soluble basic peptide complexes, pharmaceutical formulations containing them, and the use thereof as medicaments
US7098305B2 (en) * 2001-09-06 2006-08-29 Ardana Bioscience Limited Sustained release of microcrystalline peptide suspensions
US20060210639A1 (en) * 2005-03-17 2006-09-21 Elan Pharma International Limited Nanoparticulate bisphosphonate compositions
US20070021378A1 (en) * 2005-07-22 2007-01-25 The Regents Of The University Of California Heparin compositions and selectin inhibition
US20070021357A1 (en) * 2005-06-17 2007-01-25 Dynamis Therapeutics, Inc. Treatment of inflammatory conditions
US20070060509A1 (en) * 2003-12-13 2007-03-15 Venkata-Rangarao Kanikanti Endoparasiticidal compositions for topical application
US20070077313A1 (en) * 2005-10-04 2007-04-05 U.S. Pharmaceutical Corporation Toleration iron supplement compositions
US20070148228A1 (en) * 1999-02-22 2007-06-28 Merrion Research I Limited Solid oral dosage form containing an enhancer
US20070196464A1 (en) * 1999-02-22 2007-08-23 Merrion Research I Limited Solid oral dosage form containing an enhancer
US20070212395A1 (en) * 2006-03-08 2007-09-13 Allergan, Inc. Ocular therapy using sirtuin-activating agents
US20070219131A1 (en) * 2004-04-15 2007-09-20 Ben-Sasson Shmuel A Compositions capable of facilitating penetration across a biological barrier
US20080171848A1 (en) * 2004-08-31 2008-07-17 Novo Nordisk A/S Use of Tris(Hydroxymethyl) Aminomethane For the Stabilization of Peptides, Polypeptides and Proteins
US7410957B2 (en) * 2002-05-10 2008-08-12 Hoffmann-La Roche Inc. Method of treatment using bisphosphonic acid
US20080213366A1 (en) * 2005-04-29 2008-09-04 Cubist Pharmaceuticals, Inc Therapeutic Compositions
US20090004281A1 (en) * 2007-06-26 2009-01-01 Biovail Laboratories International S.R.L. Multiparticulate osmotic delivery system
US20090060861A1 (en) * 2005-05-25 2009-03-05 Novo Nordisk A/S Stabilized Polypeptide Formulations
US20100022480A1 (en) * 2006-04-07 2010-01-28 Merrion Research Iii Limited Solid Oral Dosage Form Containing An Enhancer
US7670626B2 (en) * 2001-07-02 2010-03-02 Merrion Research Iii Limited Delivery of a bioactive material
US20100105627A1 (en) * 2008-09-17 2010-04-29 Paul Salama Pharmaceutical compositions and related methods of delivery
US20120189692A1 (en) * 2011-01-07 2012-07-26 Alan Cullen Pharmaceutical Compositions of Iron for Oral Administration

Family Cites Families (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3010A (en) * 1843-03-21 Iiziiiiijiiii
US599432A (en) * 1898-02-22 Wagon-jack
GB953626A (en) 1961-04-25 1964-03-25 Meito Sangyo Kk Enteric-coated tablets of dextran sulphate ester and method of preparation thereof
JPS57146722A (en) 1981-03-06 1982-09-10 Toyo Jozo Co Ltd Pharmaceutical preparation having improved absorption
JPS5973600A (en) 1982-10-19 1984-04-25 Okayasu Shoten:Kk Ovulatory inducing agent
BR8406921A (en) 1983-05-31 1985-06-04 Choong Gook Jang DRY COMPRESSION COMPOSITES, DIRECT FOR CONTROLLED RELEASE DOSAGE FORMS
CH667994A5 (en) 1986-05-27 1988-11-30 Sandoz Ag Pharmaceutical compsns.
US4786508A (en) * 1986-05-30 1988-11-22 Warner-Lambert Company Coated dosage forms
US4789547A (en) 1987-06-17 1988-12-06 Warner-Lambert Company Transdermal matrix system
GB2212396A (en) 1987-12-18 1989-07-26 Procter & Gamble Dietary supplement comprising calcium and delayed release coated iron
NZ228285A (en) * 1988-03-11 1991-08-27 Teikoku Seiyaku Kk Pharmaceutical composition comprising a polypeptide and adapted for intravaginal administration
US5169933A (en) 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
ZA898331B (en) 1988-11-22 1990-07-25 Hoffmann La Roche Pharmaceutical compositions
IL92537A (en) 1988-12-15 1994-04-12 Riker Laboratories Inc Topical formulations and transdermal delivery systems containing- isobutyl-1H-imidazo [4,5-c] quinolin-4-amine
JP3009911B2 (en) * 1989-06-28 2000-02-14 エーザイ株式会社 Novel formulation of peptide or protein for internal use
GB2244015B (en) 1990-03-14 1993-10-27 Hadley Sections Limited Tube and method of forming same
JPH03275633A (en) 1990-03-23 1991-12-06 Teikoku Seiyaku Co Ltd Absorbefacient for physiologically active polypeptide
JPH04149126A (en) 1990-10-09 1992-05-22 Mitsubishi Kasei Corp Pharmaceutical composition for transmucous administration
IT1245761B (en) 1991-01-30 1994-10-14 Alfa Wassermann Spa PHARMACEUTICAL FORMULATIONS CONTAINING GLYCOSAMINOGLICANS ABSORBABLE ORALLY.
US5688761A (en) * 1991-04-19 1997-11-18 Lds Technologies, Inc. Convertible microemulsion formulations
AU653026B2 (en) 1991-06-07 1994-09-15 Teikoku Seiyaku Kabushiki Kaisha Physiologically active polypeptide-containing pharmaceutical composition
KR100274734B1 (en) 1991-11-22 2000-12-15 제이코버스 코넬리스 레이서 Risedronate delayed-release compositions
US5206219A (en) * 1991-11-25 1993-04-27 Applied Analytical Industries, Inc. Oral compositions of proteinaceous medicaments
SE501389C2 (en) 1992-04-24 1995-01-30 Leiras Oy Pharmaceutical preparation and process for its preparation
JP3190441B2 (en) 1992-07-20 2001-07-23 エーザイ株式会社 Stable formulation containing azelastine hydrochloride
RU2068689C1 (en) 1992-09-24 1996-11-10 Товарищество с ограниченной ответственностью "Лекрон" Method of paracetamol tablet producing
AU5376894A (en) 1992-11-06 1994-06-08 Hisamitsu Pharmaceutical Co., Inc. Peroral pharmaceutical preparation releasable in lower digestive tract
JPH06192107A (en) 1992-12-25 1994-07-12 Sanwa Kagaku Kenkyusho Co Ltd Glycyrrhizin oral agent
TW402506B (en) * 1993-06-24 2000-08-21 Astra Ab Therapeutic preparation for inhalation
US5633347A (en) * 1993-06-29 1997-05-27 University Of Utah Research Foundation Conotoxin peptides
ES2199981T3 (en) 1994-02-16 2004-03-01 Abbott Laboratories MODE OF PREPARATION OF PHARMACEUTICAL FORMULATIONS OF FINE PARTICLES.
DE69535432T2 (en) * 1994-04-22 2007-12-06 Astellas Pharma Inc. Colon-specific drug delivery system
IL110024A (en) 1994-06-15 1998-04-05 Yissum Res Dev Co Controlled release oral drug delivery system containing hydrogel- forming polymer
ES2180658T3 (en) 1994-11-17 2003-02-16 Toray Industries PREPARED FOR PERCUTANEOUS ABSORPTION.
CA2220451A1 (en) 1995-05-17 1996-11-21 Cedars-Sinai Medical Center Methods and compositions for improving digestion and absorption in the small intestine
US6572879B1 (en) 1995-06-07 2003-06-03 Alza Corporation Formulations for transdermal delivery of pergolide
ATE256456T1 (en) 1996-05-17 2004-01-15 Merck & Co Inc BIPHOSPHONATE FORMULATION WITH EFFORTABLE EFFECT
US6150522A (en) 1996-05-20 2000-11-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6147088A (en) 1996-05-20 2000-11-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
AU710926B2 (en) 1996-05-20 1999-09-30 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
GB9614235D0 (en) 1996-07-06 1996-09-04 Danbiosyst Uk Composition for enhanced uptake of polar drugs from mucosal surfaces
US6156772A (en) 1997-06-05 2000-12-05 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6156767A (en) 1997-06-05 2000-12-05 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6004984A (en) 1997-06-05 1999-12-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
IL121269A0 (en) 1997-07-09 1998-01-04 Dpharm Ltd Compositions and methods for reversibly increasing permeability of biomembranes
IL121268A0 (en) 1997-07-09 1998-01-04 Dpharm Ltd Branched chain fatty acids their derivatives and use in the treatment of central nervous system disorders
JP4181232B2 (en) 1997-07-18 2008-11-12 帝國製薬株式会社 Diclofenac sodium-containing oily external patch preparation
US6326360B1 (en) 1998-03-11 2001-12-04 Grelan Pharmaceuticals Co., Ltd. Bubbling enteric coated preparations
US5998432A (en) 1998-04-02 1999-12-07 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
GB2336311A (en) 1998-04-15 1999-10-20 Merck & Co Inc Bisphosphonate Dosing Regimen
US6656922B2 (en) 1998-05-28 2003-12-02 Mediplex Corporation, Korea Oral delivery of macromolecules
AU1071200A (en) 1998-10-19 2000-05-08 Biotech Australia Pty Limited Systems for oral delivery
DE60038097T2 (en) 1999-02-22 2009-02-12 Merrion Research I Ltd. SOLID ORAL DOSAGE FORM CONTAINING A RESORPTION AMPLIFIER
SE9901272D0 (en) 1999-04-09 1999-04-09 Astra Ab New improved formulation
JP2001081031A (en) * 1999-08-30 2001-03-27 Schering Ag Benzamide derivative-containing preparation having improved solubility and oral adsorption
DE60011678T2 (en) 1999-12-08 2005-07-14 Xcyte Therapies, Inc., Seattle DEPSIPEPTIDES AND ITS ANALOGS FOR USE AS IMMUNOSUP PRESSIVES
US6468559B1 (en) 2000-04-28 2002-10-22 Lipocine, Inc. Enteric coated formulation of bishosphonic acid compounds and associated therapeutic methods
IT1318539B1 (en) * 2000-05-26 2003-08-27 Italfarmaco Spa PROLONGED RELEASE PHARMACEUTICAL COMPOSITIONS FOR THE PARENTERAL ADMINISTRATION OF BIOLOGICALLY HYDROPHILE SUBSTANCES
GB0029111D0 (en) 2000-11-29 2001-01-10 Novartis Ag Organic compounds
EP1390048A2 (en) 2001-02-16 2004-02-25 Shimizu Pharmaceutical Co., Ltd. Oral formulations containing mucopolysaccharide for small intestine delivery and their use in the treatment circulatory disorders
US7820722B2 (en) 2001-05-11 2010-10-26 Merrion Research Iii Limited Permeation enhancers
US20030018085A1 (en) 2001-05-11 2003-01-23 Raoof Araz A. Isostearic acid salts as permeation enhancers
DE10157628A1 (en) * 2001-11-26 2003-06-12 Zentaris Ag Solution for injection of an LHRH antagonist
WO2003047493A2 (en) 2001-12-03 2003-06-12 Dor Biopharma Inc. Stabilized reverse micelle compositions and uses thereof
CN1606432A (en) 2001-12-19 2005-04-13 阿尔扎公司 Formulation and dosage form for increasing oral bioavailability of hydrophilic macromolecules
JP4259324B2 (en) * 2002-02-28 2009-04-30 ニプロ株式会社 Stabilized albumin preparation
US7154002B1 (en) 2002-10-08 2006-12-26 Takeda San Diego, Inc. Histone deacetylase inhibitors
AU2004264958B2 (en) * 2003-08-13 2010-04-15 Biocon, Ltd Micro-particle fatty acid salt solid dosage formulations for therapeutic agents
DE10345944B4 (en) * 2003-10-02 2007-06-21 Robert Bosch Gmbh Sensor for determining a physical property of a gas mixture
AR046773A1 (en) 2003-12-23 2005-12-21 Novartis Ag PHARMACEUTICAL FORMULATIONS OF BISPHOSPHONATES
US20050221501A1 (en) * 2003-12-24 2005-10-06 Arnot Kate I Dissolution method
JPWO2005072747A1 (en) 2004-02-02 2007-09-13 小野薬品工業株式会社 Bone resorption inhibitor
EP1742663A2 (en) * 2004-04-15 2007-01-17 Chiasma, Ltd. Compositions capable of facilitating penetration across a biological barrier
MXPA06013575A (en) 2004-05-24 2007-02-08 Procter & Gamble Dosage forms of bisphosphonates.
JP2006028031A (en) * 2004-07-12 2006-02-02 Ltt Bio-Pharma Co Ltd Medicine-sealed nano particle for transmucosa absorption
CA2574085C (en) * 2004-07-16 2014-02-11 Oakwood Laboratories, L.L.C. Gonadotropin releasing hormone antagonists
JP2008509933A (en) * 2004-08-13 2008-04-03 エミスフェアー・テクノロジーズ・インク Pharmaceutical formulation comprising microparticles or nanoparticles of a delivery agent
US20080125361A1 (en) * 2004-11-12 2008-05-29 Novo Nordisk A/S Stable Formulations Of Peptides
TWI362392B (en) 2005-03-18 2012-04-21 Novo Nordisk As Acylated glp-1 compounds
WO2006103657A2 (en) 2005-03-31 2006-10-05 Dexcel Pharma Technologies Ltd. A solid composition for intra-oral delivery of insulin
IL174387A0 (en) 2005-03-31 2008-01-20 Dexcel Pharma Technologies Ltd A solid composition for intra-oral delivery of insulin
AU2006241072A1 (en) * 2005-04-25 2006-11-02 Amgen Inc. Biodegradable peptide sustained release compositions containing porogens
US9283260B2 (en) * 2006-04-21 2016-03-15 Amgen Inc. Lyophilized therapeutic peptibody formulations
CA2654566A1 (en) 2006-06-09 2007-12-21 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
CN101125132A (en) * 2007-07-25 2008-02-20 司炳奎 Oral insulin enteric capsule and its preparation method
CN102046192B (en) * 2008-05-07 2014-11-19 默里昂研究Ⅲ有限公司 Compositions of GnRH related compounds and processes of preparation
CA2751854A1 (en) * 2009-02-25 2010-09-02 Merrion Research Iii Limited Composition and drug delivery of bisphosphonates
WO2011120033A1 (en) 2010-03-26 2011-09-29 Merrion Research Iii Limited Pharmaceutical compositions of selective factor xa inhibitors for oral administration

Patent Citations (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4900730A (en) * 1981-01-14 1990-02-13 Toyo Jozo Co., Ltd. Preparation which promotes the absorption of peptides
US4525339A (en) * 1982-10-15 1985-06-25 Hoffmann-La Roche Inc. Enteric coated oral dosage form
US4656161A (en) * 1983-08-27 1987-04-07 Basf Aktiengesellschaft Increasing the enteral absorbability of heparin or heparinoids
US4590062A (en) * 1984-04-16 1986-05-20 Tech Trade Corp. Dry direct compression compositions for controlled release dosage forms
US4654155A (en) * 1985-03-29 1987-03-31 Reynolds Metals Company Microemulsion lubricant
US5288497A (en) * 1985-05-01 1994-02-22 The University Of Utah Compositions of oral dissolvable medicaments
US4764375A (en) * 1985-09-11 1988-08-16 Kv Pharmaceutical Company Sachet drug delivery system
US4996058A (en) * 1987-09-18 1991-02-26 Ciba-Geigy Corporation Covered retard forms
US5221734A (en) * 1987-10-01 1993-06-22 Ciba-Geigy Corporation Process for preparing a polypeptide growth factor for milk
US5190748A (en) * 1988-11-22 1993-03-02 Hoffmann-La Roche Inc. Absorption enhancement of antibiotics
US5110606A (en) * 1990-11-13 1992-05-05 Affinity Biotech, Inc. Non-aqueous microemulsions for drug delivery
US6747125B1 (en) * 1991-03-14 2004-06-08 The Salk Institute For Biological Studies Peptide intermediates for making GnRH antagonists
US5444041A (en) * 1991-04-19 1995-08-22 Ibah, Inc. Convertible microemulsion formulations
US5646109A (en) * 1991-04-19 1997-07-08 Lds Technologies, Inc. Convertible microemulsion formulations
US5633226A (en) * 1991-04-19 1997-05-27 Lds Technologies, Inc. Convertible microemulsion formulations
US5229130A (en) * 1991-12-20 1993-07-20 Cygnus Therapeutics Systems Vegetable oil-based skin permeation enhancer compositions, and associated methods and systems
US5229103A (en) * 1992-04-30 1993-07-20 Hydrodent Laboratories, Inc. Antiplaque mouthwash concentrate
US5821222A (en) * 1992-06-11 1998-10-13 Bayer Aktiengesellschaft Cyclic depsipeptides having 18 ring atoms for combating endoparasites
WO1994008599A1 (en) * 1992-10-14 1994-04-28 The Regents Of The University Of Colorado Ion-pairing of drugs for improved efficacy and delivery
US5346701A (en) * 1993-02-22 1994-09-13 Theratech, Inc. Transmucosal delivery of macromolecular drugs
US5714477A (en) * 1993-06-18 1998-02-03 Pharmacia & Upjohn Aktiebolag Pharmaceutical composition containing heparin, heparin fragments or their derivatives in combination with glycerol esters
US5736161A (en) * 1993-07-21 1998-04-07 Lipotec S.A. Pharmaceutical preparation for improving the bioavailability of drugs which are difficult to absorb and a procedure for obtaining it
US5707648A (en) * 1993-11-17 1998-01-13 Lds Technologies, Inc. Transparent liquid for encapsulated drug delivery
US5506207A (en) * 1994-03-18 1996-04-09 The Salk Institute For Biological Studies GNRH antagonists XIII
US5541155A (en) * 1994-04-22 1996-07-30 Emisphere Technologies, Inc. Acids and acid salts and their use in delivery systems
US5639469A (en) * 1994-06-15 1997-06-17 Minnesota Mining And Manufacturing Company Transmucosal delivery system
US6017559A (en) * 1994-07-15 2000-01-25 Dow Agrosciences Llc Preparation of aqueous emulsions
US6524557B1 (en) * 1994-12-22 2003-02-25 Astrazeneca Ab Aerosol formulations of peptides and proteins
US5650386A (en) * 1995-03-31 1997-07-22 Emisphere Technologies, Inc. Compositions for oral delivery of active agents
US5631347A (en) * 1995-06-07 1997-05-20 Eli Lilly And Company Reducing gelation of a fatty acid-acylated protein
US6200602B1 (en) * 1995-08-08 2001-03-13 West Pharmaceutical Services Drug Delivery & Clinical Research Centre Limited Composition for enhanced uptake of polar drugs from the colon
US5863555A (en) * 1995-10-23 1999-01-26 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
US5807983A (en) * 1995-12-28 1998-09-15 The Salk Institute For Biological Studies GNRH antagonist betides
US6124261A (en) * 1996-07-03 2000-09-26 Alza Corporation Non-aqueous polar aprotic peptide formulations
US6235712B1 (en) * 1996-07-03 2001-05-22 Alza Corporation Non-aqueous polar aprotic peptide formulations
US6068850A (en) * 1996-07-03 2000-05-30 Alza Corporation Aqueous formulations of peptides
US5912009A (en) * 1996-10-30 1999-06-15 Theratech, Inc. Fatty acid esters of glycolic acid and its salts
US5952000A (en) * 1996-10-30 1999-09-14 Theratech, Inc. Fatty acid esters of lactic acid salts as permeation enhancers
US5821230A (en) * 1997-04-11 1998-10-13 Ferring Bv GnRH antagonist decapeptides
US6214798B1 (en) * 1997-04-11 2001-04-10 Ferring Bv GnRH antagonists being modified in positions 5 and 6
US6077858A (en) * 1997-06-05 2000-06-20 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6747014B2 (en) * 1997-07-01 2004-06-08 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
US6015801A (en) * 1997-07-22 2000-01-18 Merck & Co., Inc. Method for inhibiting bone resorption
US6372728B1 (en) * 1997-10-10 2002-04-16 Astrazeneca Ab Formulation for treatment of osteoporosis
US6017944A (en) * 1997-10-28 2000-01-25 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6077847A (en) * 1998-04-02 2000-06-20 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6025366A (en) * 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6270804B1 (en) * 1998-04-03 2001-08-07 Biovail Technologies Ltd. Sachet formulations
US6296881B1 (en) * 1998-08-03 2001-10-02 Kenkohyakunijussai Co., Ltd. Bactericide containing iron ions
US20030091623A1 (en) * 1999-02-22 2003-05-15 Cumming Kenneth Iain Solid oral dosage form containing an enhancer
US7658938B2 (en) * 1999-02-22 2010-02-09 Merrion Reasearch III Limited Solid oral dosage form containing an enhancer
US8119159B2 (en) * 1999-02-22 2012-02-21 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US20070148228A1 (en) * 1999-02-22 2007-06-28 Merrion Research I Limited Solid oral dosage form containing an enhancer
US20130089604A1 (en) * 1999-02-22 2013-04-11 Merrion Research Ill Limited Solid Oral Dosage Form Containing An Enhancer
US8828431B2 (en) * 1999-02-22 2014-09-09 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US20100028421A1 (en) * 1999-02-22 2010-02-04 Merrion Research Iii Limited Solid Oral Dosage Form Containing an Enhancer
US20070196464A1 (en) * 1999-02-22 2007-08-23 Merrion Research I Limited Solid oral dosage form containing an enhancer
US20100209499A1 (en) * 1999-02-22 2010-08-19 Cumming Kenneth I Solid Oral Dosage Form Containing an Enhancer
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US20020002140A1 (en) * 2000-01-14 2002-01-03 Holick Michael F. Novel bisphosphonates and uses thereof
US20030176397A1 (en) * 2000-04-07 2003-09-18 Lichtenberger Lenard M. Unique compositions of zwitterionic phospholipids and bisphosphonates and use of the compositions as bisphosphate delivery systems with reduced GI toxicity
US6949258B2 (en) * 2000-06-07 2005-09-27 Hao Zhang Biologically active oral preparation that can be site-specific released in colon
US20030166508A1 (en) * 2000-06-07 2003-09-04 Junshou Zhang Biologically active oral preparation that can be site-specific released in colon
US20030181421A1 (en) * 2000-06-20 2003-09-25 Horowitz Zebulun D. Method of administering bisphosphonates
US20030114525A1 (en) * 2000-11-21 2003-06-19 Kammer Gary M. Method of treating autoimmune diseases
US6379960B1 (en) * 2000-12-06 2002-04-30 Isis Pharmaceuticals, Inc. Antisense modulation of damage-specific DNA binding protein 2, p48 expression
US6875843B2 (en) * 2001-01-26 2005-04-05 Children's Mercy Hospital Prevention of diabetes and prolongation of the honeymoon phase of diabetes by administration of GnRH antagonists
US20040147484A1 (en) * 2001-03-01 2004-07-29 Boyd Maria Aurora P. Compositions for delivering bisphosphonates
US20040157799A1 (en) * 2001-05-02 2004-08-12 Seaman John J Pharmaceutical uses of bisphosphonates
US7670626B2 (en) * 2001-07-02 2010-03-02 Merrion Research Iii Limited Delivery of a bioactive material
US20030031757A1 (en) * 2001-08-03 2003-02-13 Kraft Food Holdings, Inc. Stable and bioavailable iron fortified beverages
US7098305B2 (en) * 2001-09-06 2006-08-29 Ardana Bioscience Limited Sustained release of microcrystalline peptide suspensions
US7214662B2 (en) * 2001-11-27 2007-05-08 Zentaris Gmbh Injectable solution of an LHRH antagonist
US20030100509A1 (en) * 2001-11-27 2003-05-29 Werner Sarlikiotis Injectable solution of an LHRH antagonist
US20030139378A1 (en) * 2001-12-13 2003-07-24 Daifotis Anastasia G. Liquid bisphosphonate formulations for bone disorders
US20040087631A1 (en) * 2002-03-04 2004-05-06 Bacopoulos Nicholas G. Methods of treating cancer with HDAC inhibitors
US7410957B2 (en) * 2002-05-10 2008-08-12 Hoffmann-La Roche Inc. Method of treatment using bisphosphonic acid
US20050080075A1 (en) * 2003-08-25 2005-04-14 Nichols M. James Formulations, conjugates, and combinations of drugs for the treatment of neoplasms
US20050065117A1 (en) * 2003-09-19 2005-03-24 Pfizer Inc Pharmaceutical compositions and methods comprising combinations of 2-alkylidene-19-nor-vitamin D derivatives and a bisphosphonate
US20050163849A1 (en) * 2003-10-31 2005-07-28 Wong Patrick S. Compositions and dosage forms for enhanced absorption of iron
US20050119331A1 (en) * 2003-11-04 2005-06-02 Jackie Butler Pharmaceutical formulations for carrier-mediated transport statins and uses thereof
US20070060509A1 (en) * 2003-12-13 2007-03-15 Venkata-Rangarao Kanikanti Endoparasiticidal compositions for topical application
US20050157799A1 (en) * 2004-01-15 2005-07-21 Arvind Raman System, method, and apparatus for error concealment in coded video signals
US20050249799A1 (en) * 2004-03-03 2005-11-10 Spherics, Inc. Polymeric drug delivery system for hydrophobic drugs
US20070219131A1 (en) * 2004-04-15 2007-09-20 Ben-Sasson Shmuel A Compositions capable of facilitating penetration across a biological barrier
US20060018874A1 (en) * 2004-07-19 2006-01-26 Balasingam Radhakrishnan Fatty acid formulations for oral delivery of proteins and peptides, and uses thereof
US20080171848A1 (en) * 2004-08-31 2008-07-17 Novo Nordisk A/S Use of Tris(Hydroxymethyl) Aminomethane For the Stabilization of Peptides, Polypeptides and Proteins
US20060135405A1 (en) * 2004-12-22 2006-06-22 Zentaris Gmbh Process for producing sterile suspensions of slightly soluble basic peptide complexes, sterile suspensions of slightly soluble basic peptide complexes, pharmaceutical formulations containing them, and the use thereof as medicaments
US20060210639A1 (en) * 2005-03-17 2006-09-21 Elan Pharma International Limited Nanoparticulate bisphosphonate compositions
US20080213366A1 (en) * 2005-04-29 2008-09-04 Cubist Pharmaceuticals, Inc Therapeutic Compositions
US20090060861A1 (en) * 2005-05-25 2009-03-05 Novo Nordisk A/S Stabilized Polypeptide Formulations
US20070021357A1 (en) * 2005-06-17 2007-01-25 Dynamis Therapeutics, Inc. Treatment of inflammatory conditions
US20070021378A1 (en) * 2005-07-22 2007-01-25 The Regents Of The University Of California Heparin compositions and selectin inhibition
US20070077313A1 (en) * 2005-10-04 2007-04-05 U.S. Pharmaceutical Corporation Toleration iron supplement compositions
US20070212395A1 (en) * 2006-03-08 2007-09-13 Allergan, Inc. Ocular therapy using sirtuin-activating agents
US20100022480A1 (en) * 2006-04-07 2010-01-28 Merrion Research Iii Limited Solid Oral Dosage Form Containing An Enhancer
US7704977B2 (en) * 2006-04-07 2010-04-27 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US20100247640A1 (en) * 2006-04-07 2010-09-30 Leonard Thomas W Solid Oral Dosage Form Containing An Enhancer
US20090004281A1 (en) * 2007-06-26 2009-01-01 Biovail Laboratories International S.R.L. Multiparticulate osmotic delivery system
US20100105627A1 (en) * 2008-09-17 2010-04-29 Paul Salama Pharmaceutical compositions and related methods of delivery
US20120189692A1 (en) * 2011-01-07 2012-07-26 Alan Cullen Pharmaceutical Compositions of Iron for Oral Administration

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Fattyacids_11_21_14 downloaded on 11/21/2014 *
Hovgaard, Ph. D. thesis: insulin stabilization and gastrointestinal absorption, pages1-218, publication year 1991 *
Leonard et al., Expert Opinion Drug Del. (2006), 685-692 *

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8323690B2 (en) 1999-02-22 2012-12-04 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US8119159B2 (en) 1999-02-22 2012-02-21 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US8828431B2 (en) 1999-02-22 2014-09-09 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US8323689B2 (en) 1999-02-22 2012-12-04 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US20100209499A1 (en) * 1999-02-22 2010-08-19 Cumming Kenneth I Solid Oral Dosage Form Containing an Enhancer
US8053429B2 (en) 1999-02-22 2011-11-08 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US20070196464A1 (en) * 1999-02-22 2007-08-23 Merrion Research I Limited Solid oral dosage form containing an enhancer
US20070148228A1 (en) * 1999-02-22 2007-06-28 Merrion Research I Limited Solid oral dosage form containing an enhancer
US8241670B2 (en) 2004-04-15 2012-08-14 Chiasma Inc. Compositions capable of facilitating penetration across a biological barrier
US20080159984A1 (en) * 2004-04-15 2008-07-03 Ben-Sasson Shmuel A Compositions Capable of Facilitating Penetration Across a Biological Barrier
US20070219131A1 (en) * 2004-04-15 2007-09-20 Ben-Sasson Shmuel A Compositions capable of facilitating penetration across a biological barrier
US8883203B2 (en) 2006-04-07 2014-11-11 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US8883201B2 (en) 2006-04-07 2014-11-11 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US8999383B2 (en) 2008-05-07 2015-04-07 Merrion Research Iii Limited Compositions of GnRH related compounds and processes of preparation
US9265812B2 (en) 2008-09-17 2016-02-23 Chiasma, Inc. Pharmaceutical compositions and related methods of delivery
US8535695B2 (en) 2008-09-17 2013-09-17 Chiasma Inc. Pharmaceutical compositions and related methods of delivery
US11400159B2 (en) 2008-09-17 2022-08-02 Amryt Endo, Inc. Pharmaceutical compositions and related methods of delivery
US9566246B2 (en) 2008-09-17 2017-02-14 Chiasma Inc. Pharmaceutical compositions and related methods of delivery
US8329198B2 (en) 2008-09-17 2012-12-11 Chiasma Inc. Pharmaceutical compositions and related methods of delivery
US9089484B2 (en) 2010-03-26 2015-07-28 Merrion Research Iii Limited Pharmaceutical compositions of selective factor Xa inhibitors for oral administration
US8802114B2 (en) 2011-01-07 2014-08-12 Merrion Research Iii Limited Pharmaceutical compositions of iron for oral administration
US10758592B2 (en) 2012-10-09 2020-09-01 Sanofi Exendin-4 derivatives as dual GLP1/glucagon agonists
US9745360B2 (en) 2012-12-21 2017-08-29 Sanofi Dual GLP1/GIP or trigonal GLP1/GIP/glucagon agonists
US9670261B2 (en) 2012-12-21 2017-06-06 Sanofi Functionalized exendin-4 derivatives
US10253079B2 (en) 2012-12-21 2019-04-09 Sanofi Functionalized Exendin-4 derivatives
US9751926B2 (en) 2013-12-13 2017-09-05 Sanofi Dual GLP-1/GIP receptor agonists
US9789165B2 (en) 2013-12-13 2017-10-17 Sanofi Exendin-4 peptide analogues as dual GLP-1/GIP receptor agonists
US9694053B2 (en) 2013-12-13 2017-07-04 Sanofi Dual GLP-1/glucagon receptor agonists
US9750788B2 (en) 2013-12-13 2017-09-05 Sanofi Non-acylated exendin-4 peptide analogues
US9758561B2 (en) 2014-04-07 2017-09-12 Sanofi Dual GLP-1/glucagon receptor agonists derived from exendin-4
US9771406B2 (en) 2014-04-07 2017-09-26 Sanofi Peptidic dual GLP-1/glucagon receptor agonists derived from exendin-4
US9775904B2 (en) 2014-04-07 2017-10-03 Sanofi Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
US10265384B2 (en) 2015-01-29 2019-04-23 Novo Nordisk A/S Tablets comprising GLP-1 agonist and enteric coating
US9993520B2 (en) 2015-01-30 2018-06-12 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9937223B2 (en) 2015-01-30 2018-04-10 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9919026B2 (en) 2015-01-30 2018-03-20 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9744209B2 (en) 2015-01-30 2017-08-29 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US10010575B2 (en) 2015-01-30 2018-07-03 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9962422B2 (en) 2015-01-30 2018-05-08 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9968649B2 (en) 2015-01-30 2018-05-15 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9974827B2 (en) 2015-01-30 2018-05-22 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9981006B2 (en) 2015-01-30 2018-05-29 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9375478B1 (en) 2015-01-30 2016-06-28 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9925234B2 (en) 2015-01-30 2018-03-27 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9925233B2 (en) 2015-01-30 2018-03-27 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9750785B2 (en) 2015-01-30 2017-09-05 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9744239B2 (en) 2015-01-30 2017-08-29 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US9687526B2 (en) 2015-01-30 2017-06-27 Par Pharmaceutical, Inc. Vasopressin formulations for use in treatment of hypotension
US11338011B2 (en) 2015-02-03 2022-05-24 Amryt Endo, Inc. Method of treating diseases
US11510963B1 (en) 2015-02-03 2022-11-29 Amryt Endo, Inc. Method of treating diseases
US11857595B2 (en) 2015-02-03 2024-01-02 Amryt Endo, Inc. Method of treating diseases
US10806797B2 (en) 2015-06-05 2020-10-20 Sanofi Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate
US9982029B2 (en) 2015-07-10 2018-05-29 Sanofi Exendin-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
US11890316B2 (en) 2020-12-28 2024-02-06 Amryt Endo, Inc. Oral octreotide therapy and contraceptive methods

Also Published As

Publication number Publication date
AU2009244797B2 (en) 2015-01-22
EP2288368A1 (en) 2011-03-02
CN102046192A (en) 2011-05-04
JP2016172745A (en) 2016-09-29
TW200950801A (en) 2009-12-16
RU2010150093A (en) 2012-06-20
US20090280170A1 (en) 2009-11-12
IL209136A (en) 2015-08-31
HK1157185A1 (en) 2012-06-29
CN102056615A (en) 2011-05-11
EP2288369B1 (en) 2014-07-09
CA2723558A1 (en) 2009-11-12
KR20110007242A (en) 2011-01-21
AU2009244799A1 (en) 2009-11-12
CN102046192B (en) 2014-11-19
TW200950799A (en) 2009-12-16
JP2015155433A (en) 2015-08-27
AU2009244799B2 (en) 2014-11-20
EP2288369A1 (en) 2011-03-02
AU2009244797A1 (en) 2009-11-12
BRPI0912384A2 (en) 2015-10-13
JP2011519928A (en) 2011-07-14
JP5671451B2 (en) 2015-02-18
CA2723541A1 (en) 2009-11-12
WO2009137080A1 (en) 2009-11-12
US20150238561A1 (en) 2015-08-27
JP2011519929A (en) 2011-07-14
IL209136A0 (en) 2011-01-31
MX2010012201A (en) 2011-05-30
WO2009137078A1 (en) 2009-11-12
KR20110007614A (en) 2011-01-24
US8999383B2 (en) 2015-04-07
RU2517135C2 (en) 2014-05-27
CN107096012A (en) 2017-08-29

Similar Documents

Publication Publication Date Title
AU2009244797B2 (en) Compositions of peptides and processes of preparation thereof
CN103764159A (en) Aromatic-cationic peptides and uses of same
KR102294577B1 (en) solid oral dosage form
US20150297726A1 (en) Sustained-release lipid pre-concentrate of gnrh analogues and pharmaceutical composition comprising the same
CZ337198A3 (en) Peptide pharmaceutical products for peroral application
AU2009321745A1 (en) Oral dosage forms of bendamustine
WO2016004067A1 (en) Aromatic-cationic peptide formulations, compositions and methods of use
WO2015162195A1 (en) Fatty acid acylated amino acids for oral peptide delivery
US20240041983A1 (en) Improved pharmaceutical formulations of glp-1 receptor agonists
CN104884078A (en) Fatty acid acylated D-amino acids for oral peptide delivery
KR102216578B1 (en) Oral pharmaceutical composition comprising teriparatide and method for preparing the same
JP2007535545A (en) Formulation for oral administration of poorly absorbable drugs
JP4599714B2 (en) Oral absorption improving pharmaceutical composition
EA012982B1 (en) Stabilized leukotriene b4 (ltb4) agent pharmaceutical
CN104888226A (en) Protein and/or polypeptide substance preparation
JPWO2003059389A1 (en) Pharmaceutical composition for improving oral absorption

Legal Events

Date Code Title Description
AS Assignment

Owner name: MERRION RESEARCH III LIMITED, IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEONARD, THOMAS W.;REEL/FRAME:022816/0214

Effective date: 20090528

AS Assignment

Owner name: NOVO NORDISK A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MERRION RESEARCH III LIMITED;REEL/FRAME:038292/0131

Effective date: 20160322

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION