WO2012140155A1 - Fatty acid acylated amino acids for oral peptide delivery - Google Patents

Fatty acid acylated amino acids for oral peptide delivery Download PDF

Info

Publication number
WO2012140155A1
WO2012140155A1 PCT/EP2012/056708 EP2012056708W WO2012140155A1 WO 2012140155 A1 WO2012140155 A1 WO 2012140155A1 EP 2012056708 W EP2012056708 W EP 2012056708W WO 2012140155 A1 WO2012140155 A1 WO 2012140155A1
Authority
WO
WIPO (PCT)
Prior art keywords
sodium
decanoyl
dodecanoyl
amino acid
capric
Prior art date
Application number
PCT/EP2012/056708
Other languages
French (fr)
Inventor
Florian Anders FÖGER
Abdallah MAKHLOF
Herbert Hoyer
Original Assignee
Novo Nordisk A/S
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 Novo Nordisk A/S filed Critical Novo Nordisk A/S
Priority to EP12713173.8A priority Critical patent/EP2696847A1/en
Priority to US14/111,274 priority patent/US20140056953A1/en
Priority to CN201280017853.5A priority patent/CN103458873B/en
Priority to JP2014504327A priority patent/JP6030630B2/en
Publication of WO2012140155A1 publication Critical patent/WO2012140155A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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

Definitions

  • the technical field of this invention relates to fatty acid acylated amino acids (FA- aa's) for oral delivery of therapeutic hydropilic peptides and proteins and pharmaceutical compositions comprising such FA-aa's.
  • FA- aa's fatty acid acylated amino acids
  • a non-limiting example of a hydrophilic proteins and polypeptides is human insulin which ais degraded by various digestive enzymes found in the stomach (pepsin), in the intestinal lumen (chymotrypsin, trypsin, elastase, carboxypeptidases, etc.) and in the mucosal surfaces of the Gl tract (aminopeptidases, carboxypeptidases, enteropeptidases, dipeptidyl peptidases, endopeptidases, etc.).
  • WO2004147578 relates to fatty acid acylated amino acids used as permeation enhancers for hydrophobic molecules including hydrophobic macromolecules such as cyclosporine.
  • WO2001035998 relates to acylated amino acids used as transdermal and transmucosal absorption promoters for macromolecules, such as hydrophilic peptides or proteins.
  • WO2004064758 relates to an oral composition for delivering pharmaceutical peptides, such as insulin, growth hormone and GLP-1 , comprising absorption enhancers, including acyl amino acids.
  • US2005282756 is related to a dry powder composition comprising insulin and an absorption enhancer.
  • WO2003030865 is related to insulin compositions comprising surfactants such as ionic surfactants and does also contain oil or lipid compounds such as triglycerides and does further comprise long chain esterified fatty acids (C12 to C18).
  • WO2004064758 is related to an oral pharmaceutical composition for delivering pharmaceutical peptides, comprising absorption enhancers.
  • the oral route of administration is rather complex and a need for establishment of an acceptable composition suitable for the treatment of patients, with an effective
  • bioavailability of the macromolecule such as hydrophilic peptides or proteins, is existent.
  • This invention is an oral pharmaceutical composition
  • an oral pharmaceutical composition comprising certain amino acids acylated at their alpha-amino group with a fatty acid of 8 to 18 carbons and and an active ingredient, such as a hydrophilic peptide or protein.
  • the fatty acid chain distribution in the cocoyl sarcosinate is 1 % C6, 8% C8, 6% C10, 48% C12, 18% C14, 8% C16, 6% C18 saturated and 5% C18 unsaturated.
  • the fatty acid chain distribution in the cocoyl sarcosinate is 1 % C6, 8% C8, 6% C10, 48% C12, 18% C14, 8% C16, 6% C18 saturated and 5% C18 unsaturated.
  • B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of different fatty acid acylated amino acids after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n 5-6).
  • B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in propylene glycol in presence of sodium N-capric leucine after injection into mid- jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n 6).
  • B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of different fatty acid acylated amino acids or mixtures thereof according to the invention or in presence of commonly used permeation enhancers after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n 5-6).
  • B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg) dissolved in liquid SEDDS, SMEDDS and SNEDDS formulations comprising sodium N- lauroyl phenylalanine after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n 5-6).
  • the compositions are shown in table 1 .
  • FIG. 14 Pharmakokinetic profile in a single beagle dog is shown of the insulin derivative A1 (N,N-Dimethyl), A14E, B1 (N, N-dimethyl), B25H, B29K(N(eps)octadecanedioyl-gGlu- OEG-OEG), desB30 human insulin (120 nmol/kg) after peroral dosing of an enteric coated soft capsule comprising 30mg of sodium lauroyl leucine sodium salt, 150mg of propylene glycol, 300mg of Polysorbate 20 and 520mg of diglycerol monocaprylate.
  • the present invention is related to pharmaceutical compositions, comprising FA-aa's acting as permeation enhancers suitable for oral administration of therapeutic agents
  • macromolecules e.i. therapeutic acvtive peptides and proteins. More specifically therapeutic macromolecules, such as hydrophilic peptides or proteins according to the present invention are hydrophilic peptides and proteins which have a therapeutical activity and include but are not limited to insulin.
  • hydrophilic peptides or proteins which have a therapeutical activity and include but are not limited to insulin.
  • FA-aa's are amino acid based surfactants and thus mild biodegradable surfactants with a low toxicity.
  • FA-aa's according to the present invention are valuable ingredients in oral pharmaceutical compositions. Especially valuable are FA-aa's according to this invention in oral pharmaceutical compositions comprising hydrophilic peptides or proteins as active ingredient. This is of interest for diseases that demand chronic administration of therapeutic macromolecules (e.g. peptides or proteins), but is not limited hereto, since the most non-invasive, non-toxic administration of drugs is generally favoured in any treatment, also for sporadic or bulk administration of therapeutics.
  • therapeutic macromolecules e.g. peptides or proteins
  • the invention may also solve further problems that will be apparent from the disclosure of the exemplary embodiments.
  • the present invention is related to oral pharmaceutical compositions comprising FA-aa's suitable for increasing the bioavailability of therapeutic macromolecules (e.g. peptides and proteins) and their absorption.
  • therapeutic macromolecules e.g. peptides and proteins
  • One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule, such as hydrophilic peptides or proteins and at least one FA-aa.
  • One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is ahydrophilic peptide or protein.
  • This invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is a peptide.
  • This invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is a therapeutic active peptide.
  • One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is a protein.
  • One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is a therapeutic protein.
  • One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is an insulin.
  • One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is an insulin peptide.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcos
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcos
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcos
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcos
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid
  • said nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a therapeutic macromolecule such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms
  • said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a polar uncharged amino acid.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a polar uncharged amino acid
  • said polar uncharged amino acid may be selected from the group consisting of Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a polar acidic amino acid.
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a polar acidic amino acid, said polar acidic amino acid may be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and FA-aa's, based on a mixture FA-aa's.
  • a pharmaceutical composition according to the present invention comprises one or more commercially available FA-aa's.
  • a FA-aa comprises an amino residue and a fatty acid attached to the amino acid by acylation of said amino acid's alpha-amino group.
  • an amino acid residue according to this invention includes the form of its free acid or a salt.
  • an amino acid residue according to this invention includes the form of its free acid or sodium (Na+) salt.
  • a FA-aa comprises an acylated amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms.
  • a FA-aa comprises an acylated amino acid and a fatty acid moiety consisting of 10 carbon atoms.
  • a FA-aa comprises an acylated amino acid and a fatty acid moiety consisting of 12 carbon atoms.
  • a FA-aa comprises an acylated amino acid and a fatty acid moiety consisting of 14 carbon atoms.
  • a FA-aa comprises an acylated amino acid and a fatty acid moiety consisting of 16 carbon atoms.
  • a FA-aa comprises an acylated amino acid and a fatty acid moiety consisting of 18 carbon atoms.
  • a FA-aa comprises an amino acid residue acylated with a fatty acid or salt thereof.
  • a FA-aa comprises an acylated amino acid, wherein the fatty acid moiety is located at the alpha amino group of the amino acid.
  • a FA-aa comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 8 to 18 carbon atoms.
  • a FA-aa comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 10 carbon atoms.
  • a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 12 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 14 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 16 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 18 carbon atoms.
  • a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is in the form of its free acid or salt. In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is in the form of its free acid or sodium (Na+) salt. In one embodiment a FA-aa according to this invention comprise amino acid residues in the form of their free acid or a salt. In one embodiment a FA-aa according to this invention comprises amino acid residues in the form of their free acid or a sodium (Na+) salt. In one embodiment a FA-aa according to this invention is soluble at intestinal pH values, particularly in the 5.5 to 8.0 range. In one embodiment a FA-aa according to this invention is soluble at intestinal pH values, particularly in the 6.5 to 7.0 range.
  • a FA-aa according to this invention has a solubility of at least 10mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 20mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 30mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 40 mg/mL. In one embodiment a FA-aa according to this invention has a solubility of at least 50mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 60mg/ml_.
  • a FA-aa according to this invention has a solubility of at least 70mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 80mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 90mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 100mg/ml_.
  • a FA-aa according to this invention has a solubility of at least 5mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 10mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 20mg/ml_in water. In one embodiment a FA-aa according to this invention has a solubility of at least 30mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 40 mg/mL in water. In one embodiment a FA-aa according to this invention has a solubility of at least 50mg/ml_ in water.
  • a FA-aa according to this invention has a solubility of at least 60mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 70mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 80mg/ml_in water. In one embodiment a FA-aa according to this invention has a solubility of at least 90mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 100mg/ml_ in water.
  • a FA-aa according to the present invention may be represented by the general formula A-X, wherein A is an amino acid residue, based on a non-cationic amino acid and X is a fatty acid attached by acylation to A's alpha amino group.
  • a FA-aa may be represented by the general formula A-X, wherein A is an amino acid residue, based on a nonpolar hydrophobic amino acid and X is a fatty acid attached by acylation to A's alpha amino group.
  • a FA-aa may be represented by the general formula A-X, wherein A is an amino acid residue, based on a polar uncharges amino acid and X is a fatty acid attached by acylation to As alpha amino group.
  • a FA-aa may be represented by the general formula A-X, wherein A is an amino acid residue, based on a polar acidic amino acid and X is a fatty acid attached by acylation to As alpha amino group.
  • a FA-aa according to the present invention may be represented by the general formula
  • R1 is a fatty acid chain comprising between from 8 to 18 carbons
  • R2 is either H (i.e. hydrogen) or CH 3 (i.e. methyl group)
  • R3 is either H, or a salt thereof
  • R4 is an amino acid side chain of a non-cationic amino acid.
  • R1 is a fatty acid chain comprising 8 to 18 carbon atoms
  • R2 is either H (i.e.
  • R3 is either H, or a sodium salt (Na + ) thereof, and R4 is a amino acid side chain of a non-cationic amino acid.
  • a FA-aa may be chosen from the group consisting of formula (a), (b), (c), (d), (e), (f), (g), (h), (i), (j), (k), (I), (m), (n), (o), (p), (q) or (r), wherein R1 is a fatty acid chain comprising between from 8 to 18 carbons, R2 is either H (i.e. hydrogen) or CH 3 (i.e. methyl group), and R3 is either H, or a salt thereof.
  • a FA-aa may be chosen from the group consisting of formula (a), (b), (c), (d), (e), (f), (g), (h), (i), (j), (k), (I), (m), (n), (o), (p), (q) or (r), wherein R1 is a fatty acid chain comprising 8 to 18 carbon atoms, R2 is either H (i.e. hydrogen) or CH 3 (i.e. methyl group), and R3 is either H, or a sodium (Na+) salt thereof.
  • amino acid residue according to this invention may be based on a non-cationic amino acid.
  • An amino acid residue according to this invention may be based on a non-cationic amino acid, said non-cationic amino acid may be selected from the group consisting nonpolar hydrophobic amino acids, polar uncharged amio acids and polar acidic amino acids.
  • An amino acid residue according to this invention may be based on a non-cationic amino acid
  • said non-cationic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
  • an amino acid residue of a FA-aa according to this invention may be based on a nonpolar hydrophobic amino acid.
  • an amino acid residue of a FA-aa may be based on a nonpolar hydrophobic amino acid
  • said nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • a FA-aa comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms
  • said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • a FA-aa comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms.
  • a FA-aa comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms
  • said nonpolar hydrophobic amino acid can be selected from the group consisting Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • a FA-aa can be selected from the group consisting of: Sodium capric alaninate, N-decanoyl-L-alanine, Sodium capric isoleucinate, N-decanoyl-L-isoleucine, Sodium capric leucinate, N-decanoyl-L-leucine, Sodium capric methioninate, N-decanoyl-L- methionine, Sodium capric phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric prolinate, N-decanoyl-L-proline, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L-tryptophane, Sodium capric valinate, N-decanoyl-L- valine, Sodium capric sarcosinate and N-decanoyl-L
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms.
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms
  • said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
  • a FA-aa can be selected from the group consisting of: Sodium lauroyl alaninate, N-dodecanoyl-L-alanine, Sodium lauroyl isoleucinate, N-dodecanoyl-L- isoleucine, Sodium lauroyl leucinate, N-dodecanoyl-L-leucine, , Sodium lauroyl methioninate, N-dodecanoyl-L-methionine, Sodium lauroyl phenylalaninate, N-dodecanoyl-L-phenylalanine, Sodium lauroyl prolinate, N-dodecanoyl-L-proline, Sodium lauroyl tryptophanate, N- dodecanoyl-L-tryptophane, Sodium lauroyl valinate, N-dodecanoyl-L-valine, Sodium lauroyl sarcos
  • a FA-aa comprises an acylated amino acid based on a nonpolar hydrophobic amino acid
  • said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a FA-aa comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms
  • said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms.
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a FA-aa can be selected from the group consisting of: Sodium capric alaninate, N-decanoyl-L-alanine, Sodium capric leucinate, N-decanoyl-L-leucine, Sodium capric phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric valinate, N- decanoyl-L-valine, Sodium N-decyl leucine,
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms.
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms.
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms.
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms.
  • a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
  • a FA-aa can be selected from the group consisting of: Sodium lauroyl alaninate, N-dodecanoyl-L-alanine, Sodium lauroyl leucinate, N-dodecanoyl-L- leucine, Sodium lauroyl phenylalaninate, N-dodecanoyl-L-phenylalanine, Sodium lauroyl valinate, N-dodecanoyl-L-valine,
  • an amino acid residue of a FA-aa according to this invention can be based on a polar uncharged amino acid.
  • an amino acid residue of a FA-aa can be based on a polar uncharged amino acid, said polar uncharged amino acid can be selected from the group consisting of Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin).
  • a FA-aa can be selected from the group consisting of: Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L- aspartic acid, Sodium lauroyl cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyl glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyl glycinate, N-dodecanoyl-L-glycine, Sodium lauroyl serinate, N-dodecanoyl-L-serine, Sodium lauroyl threoninate, N-dodecanoyl- L-threonine, Sodium lauroyl tyrosinate, N-dodecanoyl-L-tyrosine, Sodium capric asparaginate, N-decanoyl as
  • an amino acid residue of a FA-aa according to this invention can be based on a polar acidic amino acid.
  • an amino acid residue of a FA-aa according to this invention can be based on a polar acidic amino acid, said polar acidic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • Asp Aspartic acid
  • Glu Glutamic acid
  • a FA-aa comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • a FA-aa comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • a FA-aa comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • a FA-aa comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • a FA-aa can be selected from the group consisting of: Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N- dodecanoyl-L-aspartic acid, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N- decanoyl-L-aspartic acid, Sodium capric glutamic acid and N-decanoyl-L-glutamic acid.
  • a FA-aa can be selected from the group consisting of: Amisoft HS-1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium CocoyI Glutamate) and Sodium cocoyl glutamate, Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric glutamic acid and N-decanoyl-L-glutamic acid
  • a FA-aa can be selected from the group consisting of: Amisoft HS-1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)),
  • Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium Cocoyl Glutamate) and Sodium cocoyl glutamate.
  • an amino acid residue of a FA-aa according to this invention may be based on a polar acidic amino acid.
  • an amino acid residue of a FA-aa may be based on a polar acidic amino acid, said polar acidic amino acid may be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • a FA-aa comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said nonpolar hydrophobic amino acid may be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • a FA-aa comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said nonpolar hydrophobic amino acid may be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • a FA-aa comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 18 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
  • a FA-aa may be selected from the group consisting of: Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N- dodecanoyl-L-aspartic acid, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N- decanoyl-L-aspartic acid, Sodium capric glutamic acid and N-decanoyl-L-glutamic acid.
  • an amino acid amino acid may be selected from the group constsiting of Amisoft HS-1 1 P (Sodium StearoyI Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium Cocoyl Glutamate) and Sodium cocoyl glutamate.
  • an amino acid amino acid FA-aa may be selected from the group consisting of: Sodium lauroyl asparaginate, N-dodecanoyl-L- asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium capric asparaginate, N-decanoyl-L- asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric glutamic acid and N-decanoyl-L-glutamic acid.
  • an amino acid amino acid may be selected from the group constsiting of Amisoft HS-1 1 P (Sodium StearoyI Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium Cocoyl Glutamate) and Sodium cocoyl glutamate.
  • an amino acid moiety of a FA-aa according to this invention is an amino acids that are not encoded by the genetic code.
  • an amino acid moiety of a FA-aa according to this invention is Sarcosinate.
  • an amino acid residue of a FA-aa according to this invention is an free acid or salt form of an amino acid that are not encoded by the genetic code.
  • an amino acid residue of a FA-aa according to this invention is the free acid or salt form of Sarcosinate.
  • an amino acid moiety of a FA-aa according to this invention is selected from the group comprising Leucine and Phenylalanine.
  • the FA-aa may be part of an oral pharmaceutical composition.
  • the pharmaceutical composition comprises of at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and at least on FA-aa and propylene glycol.
  • amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system, such as SEDDS,
  • the amino acid FA-aa may be used in a solid surfactant based delivery system. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system, such as SEDDS, SMEDDS or SNEDDS.
  • Liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention may be encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form.
  • solid refers to liquid compositions encapsulated in a soft or hard capsule technology, but also to tablets and multiparticulates.
  • Liquid or semisolid SEDDS, SMEDDS or SNEDDS according to the invention may be encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form which may further comprise enteric or delayed release coatings.
  • Liquid or semisolid SEDDS, SMEDDS or SNEDDS comprising FA-aa's according to the invention may be encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form which may further comprise enteric or delayed release coatings, such as poly(meth)acrylates, commercially known as Eudragit®.
  • the pharmaceutical composition is a SEDDS
  • SMEDDS or SNEDDS comprising at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and at least one FA-aa, propylene glycol.
  • the pharmaceutical composition according to the present comprises less than 10% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 9% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 8% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 7% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 6% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 5% (w/w) water. In one
  • the pharmaceutical composition according to the present comprises less than 4% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 3% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 2% (w/w) water. In one
  • the pharmaceutical composition according to the present comprises less than 1 % (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 0% (w/w) water.
  • the pharmaceutical composition according to the present invention is is a liquid. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 10% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 9% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 8% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 7% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 6% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 5% (w/w) water.
  • the pharmaceutical composition according to the present invention is is a liquid and comprises less than 4% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 3% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 2% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 1 % (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 0% (w/w) water.
  • the pharmaceutical composition comprises at least one therapeutic macromoecule.
  • a therapeutic macromolecule such as a hydrophilic peptide or protein according to this invention is a therapeutic active peptide or protein.
  • a therapeutic peptide or protein according to this invention is a hydrophilic peptide or protein.
  • hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 50mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least
  • a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 70mg/ml_ in water In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 80mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 90mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 100mg/ml_ in water.
  • a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 1 10mg/mL in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 120mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 130mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 140mg/ml_ in water.
  • a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 150/mL in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 160mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 170mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 180mg/ml_ in water.
  • a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 190mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 200mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 210mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 220mg/ml_ in water.
  • hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 230mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 240mg/ml_ in water.
  • a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 1500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 1750Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 2000Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 2250Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 2500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 2750Da.
  • a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 3000Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 3250Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 3500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 3750Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 4000Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 4250Da.
  • a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 4500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 4750Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 5000Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 1500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of between 1500Da and 5000Da.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent .
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the solvent is selected from the group consisting of water and propylene glycol.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, wherein said Polyethylene glycol sorbitan fatty acid ester is selected from the group consisting of Tween 20, Tween 21 , Tween 40, Tween 60, Tween 65, Tween 80, Tween 81 and Tween 85.
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, wherein said Polyethylene glycol sorbitan fatty acid ester is selected from the group consisting of Tween 20, Tween 21 , Tween 40, Tween 60, Tween 65, Tween 80, Tween 81 and Tween 85.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent, wherein the solvent is selected from the group consistin g of water and propylene glycol.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the Polyethylene glycol sorbitan fatty acid ester is a Polyethylene glycol sorbitan trioleate, commercially known as Tween 85.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the Polyethylene glycol sorbitan fatty acid ester is a Polyethylene glycol sorbitan trioleate, commercially known as Tween 85 and the solvent is selected form the group consisting of water and propylene glycol.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan trioleate commercially known as Tween 85 and a polar or semipolar solvent selected from the group consistin of water and propylene glycol, wherein the composition forms a microemulsion after dilution in an aqeous medium.
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan trioleate commercially known as Tween 85 and a polar or semipolar solvent selected from the group consistin of water and propylene glycol, wherein the composition forms a microemulsion after dilution in an aqeous medium.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the Polyethylene glycol sorbitan fatty acid ester is a Polyethylene glycol sorbitan trioleate, commercially known as Tween 20.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the Polyethylene glycol sorbitan fatty acid ester is a Polyethylene glycol sorbitan monolaurate, commercially known as Tween 20 and the solvent is selected form the group consisting of water and propylene glycol.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan monolaurate commercially known as Tween 20 and a polar or semipolar solvent selected from the group consistin of water and propylene glycol, wherein the composition forms a microemulsion after dilution in an aqeous medium.
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan monolaurate commercially known as Tween 20 and a polar or semipolar solvent selected from the group consistin of water and propylene glycol, wherein the composition forms a microemulsion after dilution in an aqeous medium.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent.
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising
  • Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent, wherein said polar or semipolar solvent is selected from the group consisting of water and propylene glycol.
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent, wherein said polar or semipolar solvent is selected from the group consisting of water and propylene glycol.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester and a polar or semipolar solvent (such as water or propylene glycol).
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester (Span 10, 20, 40, 60 or 80), and a polar or semipolar solvent (such as water or propylene glycol).
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester, wherein said sorbitan fatty acid ester is selected form the group consisting of Span 10, Span 20, Span 40, Span 60 and Span 80.
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester, wherein said sorbitan fatty acid ester is selected form the group consisting of sorbitan laurate commercially known as Span 20, sorbitan mono palmitate commercially known as Span 40, sorbitan mono stearate commercially known as Span 60 and sorbitan oleate commercially known as Span 80.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester and a polar or semipolar solvent.
  • a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester and a polar or semipolar solvent.
  • a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester and a polar or semipolar solvent, wherein said polar or semipolar solvent is selected from the group consisting of water or propylene glycol.
  • the pharmaceutical composition comprises at least one therapeutic active peptide or protein.
  • the pharmaceutical composition comprises at least one therapeutic active peptide or protein, which has been pH neutralised.
  • the therapeutical active peptide or protein is dissolved and the pH of the resulting solution is adjusted to a value of the target pH value, which is 1 unit, alternatively 2 units and alternatively 2.5 pH units above or below the pi of the insulin peptide, whereafter said resulting solution is freeze or spray dryed.
  • the target pH value which is 1 unit, alternatively 2 units and alternatively 2.5 pH units above or below the pi of the insulin peptide, whereafter said resulting solution is freeze or spray dryed.
  • said pH adjustment is performed with a non-volitale acid or base.
  • the pharmaceutical composition comprises of at least one insulin peptide and at least on FA-aa. In one embodiment of the invention the pharmaceutical composition comprises of at least one peptide or protein and at least on FA- aa.
  • the pharmaceutical composition comprises of at least one insulin peptide and at least on FA-aa and propylene glycol.
  • the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 10% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 9% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 8% (w/w) water.
  • the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 7% (w/w) water. In one embodiment the amino acid FA- aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 6% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 5% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 4% (w/w) water.
  • the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 3% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 2% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 1 % (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 0% (w/w) water.
  • a pharmaceutical composition according to the present invention comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one high HLB surfactant, at least one low HLB co-surfactant and a polar solvent.
  • a pharmaceutical composition according to the present invention comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one high HLB surfactant, at least one low HLB co- surfactant and a polar solvent.
  • a pharmaceutical composition according to the present invention comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least two high HLB surfactants, and a polar solvent. In one embodiment a pharmaceutical composition according to the present invention comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least two high HLB surfactants, and a polar solvent.
  • the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system, such as SEDDS, SMEDDS or SNEDDS. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 10% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 9% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 8% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 7% (w/w) water.
  • a solid surfactant based delivery system comprising less than 10% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 9% (w/w) water. In one embodiment the
  • the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 6% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 6% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 5% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 4% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 3% (w/w) water.
  • the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 2% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 1 % (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 0% (w/w) water.
  • amino acid FA-aa may be used in a solid surfactant based delivery system, such as SEDDS, SMEDDS or SNEDDS.
  • the pharmaceutical composition according to the present invention is a liquid.
  • pharmaceutical composition is a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form.
  • a soft capsule technology used for encapsulating a composition according to the present invention is gelatine free.
  • a gelatine free soft capsule technology as commercially known under the name Vegicaps® from Catalent® is used for encapsulation of the pharmaceutical composition according to the present invention.
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 10% (w/w) water.
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 9% (w/w) water
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 8% (w/w) water.
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 6% (w/w) water.
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 5% (w/w) water.
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 4% (w/w) water.
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 3% (w/w) water.
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 2% (w/w) water.
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 1 % (w/w) water
  • the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 0% (w/w) water.
  • a liquid or semisolid formulation according to the invention is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form further comprising an enteric or delayed release coatings.
  • a liquid or semisolid formulation according to the invention is encapsulated with any available enteric soft- or hard capsule technology to result in a solid oral pharmaceutical dosage.
  • a liquid or semisolid SEDDS, SMEDDS or SNEDDS In one embodiment a liquid or semisolid SEDDS, SMEDDS or SNEDDS
  • a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form further comprising an enteric or delayed release coatings.
  • a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention is
  • a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprising FA-aa's according to the invention is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form which may further comprise an enteric or delayed release coatings, such as poly(meth)acrylates, commercially known as Eudragit®.
  • the coating comprises at least one release modifying polymer which can be used to control the site where the drug (insulin derivative) is released.
  • the modified release polymer can be a polymethacrylate polymer such as those sold under the Eudragit® trade name (Evonik Rohm GmbH, Darmstadt, Germany), for example Eudragit® L30 D55, Eudragit® L100-55, Eudragit® L100, Eudragit® S100, Eudragit® S12.5,
  • Eudragit® FS30D Eudragit® NE30D and mixtures thereof as e.g. described in Eudragit® Application Guidelines, Evonik Industries, 1 1 th edition, 09/2009.
  • the pharmaceutical composition is a formulation, comprising at least one insulin and at least one FA-aa, propylene glycol.
  • the pharmaceutical composition comprises of at least one insulin and at least one FA-aa, propylene glycol.
  • the pharmaceutical comprises at least one peptide or protein and at least one FA-aa, propylene glycol.
  • the pharmaceutical composition is a SEDDS, SMEDDS or SNEDDS, comprising at least one peptide or protein and at least one FA-aa, propylene glycol.
  • the components of the drug delivery system may be present in any relative amounts.
  • the drug delivery system comprises up to 90% of a surfactant, or up to 90% of a polar organic solvent such as Polyethylene glycol (PEG) 300 g/mol, PEG 400 g/mol, PEG 600 g/mol, PEG 1000 g/mol, or up to 90% of a lipid component.
  • PEGs are prepared by polymerization of ethylene oxide and are commercially available over a wide range of molecular weights from 300 g/mol to 10,000,000 g/mol.
  • the oral pharmaceutical composition comprises from 5 to 20% of propylene glycol.
  • the oral pharmaceutical composition comprises at least one FA- aa, propylene glycol, and at least two non ionic surfactants.
  • the oral pharmaceutical composition comprises at least one FA- aa, propylene glycol, polysorbate 20 and a co-surfactant.
  • Polysorbate 20 is a polysorbate surfactant whose stability and relative non-toxicity allows it to be used as a detergent and emulsifier in a number of domestic, scientific, and pharmacological applications.
  • the number 20 refers to the total number of oxyethylene -(CH 2 CH 2 0)- groups found in the molecule.
  • the oral pharmaceutical composition comprises at least one FA-aa, propylene glycol, polysorbate 20 and a polyglycerol fatty acid ester.
  • the oral pharmaceutical composition comprises at least one FA- aa, propylene glycol, polysorbate 20 and a co-surfactant.
  • the oral pharmaceutical composition comprises at least one FA- aa, propylene glycol, polysorbate 20 and a polyglycerol fatty acid ester such as diglycerol monocaprylate.
  • the pharmaceutical composition may comprise additional excipients commonly found in pharmaceutical compositions, examples of such excipients include, but are not limited to, antioxidants, antimicrobial agents, enzyme inhibitors, stabilizers, preservatives, flavors, sweeteners and other components as described in Handbook of Pharmaceutical Excipients, Rowe et al., Eds., 4th Edition,
  • Additional excipients may be in an amount from about 0.05-5% by weight of the total pharmaceutical composition.
  • Antioxidants, anti-microbial agents, enzyme inhibitors, stabilizers or preservatives typically provide up to about 0.05-1 % by weight of the total pharmaceutical composition.
  • Sweetening or flavouring agents typically provide up to about 2.5% or 5% by weight of the total pharmaceutical composition.
  • Oral pharmaceutical compositions according to this invention may be formulated as solid dosage forms.
  • Oral pharmaceutical compositions according to this invention may be formulated as solid dosage forms and may be selected from the group consisting of capsules, tablets, dragees, pills, lozenges, powders and granules. Oral pharmaceutical compositions according to this invention may be formulated as mutliparticulate dosage forms.
  • Oral pharmaceutical compositions according to this invention may be formulated as mutliparticulate dosage forms and may be selected from the group consisting of pellets, microparticles, nanoparticles, liquid or semisolid fill formulations in soft- or hard capsules, enteric coated soft- hard capsules.
  • the oral pharmaceutical compositions may be prepared with one or more coatings such as enteric coatings or be formulated as delayed release formulations according to methods well known in the art.
  • Enteric or delayed release coatings according to this invention may be based on
  • poly(meth)acrylates commercially known as Eudragit®.
  • the pharmaceutical composition according to the invention is used for the preparation of a medicament.
  • the pharmaceutical composition according to the invention is used for the preparation of a medicament for the treatment or prevention of hyperglycemia, type 2 diabetes mellitus, impaired glucose tolerance, type 1 diabetes mellitus and/or anti obesity treatment.
  • fatty acid N-acylated amino acid or “acylated amino acid” may be used interchangeable and refer when used herein to an amino acids that is acylated with a fatty acid at its alpha-amino group.
  • Amino acids exist in the stereoisomeric form of either D (dextro) or L (levo).
  • the D and L refer to the absolute confirmation of optically active compounds.
  • all other amino acids are mirror images that can not be superimposed.
  • Most of the amino acids found in nature are of the L-type.
  • eukaryotic proteins are always composed of L-amino acids although D-amino acids are found in bacterial cell walls and in some peptide antibiotics.
  • At least 300 amino acids have been described in nature but only twenty of these are typically found as components in human peptides and proteins. Twenty standards amino acids are used by cells in peptide biosynthesis, and these are specified by the general genetic code.
  • the twenty standard amino acids are Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Methionine (Met), Proline (Pro), Apartic acid (Asp), Gltamic acid (Glu), Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Apsaragine (Asn), Glutamine (Gin), Lysine (Lys), Arginine (Arg) and Histidine (His).
  • the amino acid moiety of the modified FA-aa may be in the form of a pure enantiomer wherein the stereo configuration of the chiral amino acid moiety is either D or L (or if using the R/S terminology: either R or S) or it may be in the form of a mixture of enantiomers (D and L / R and S).
  • amino acid moiety is in the form of a mixture of enantiomers.
  • the amino moiety is in the form of a pure enantiomer. In one embodiment the chiral amino acid moiety is in the L form. In one embodiment the chiral amino acid moiety is in the D form.
  • non-cationic amino acid shall be understood as referring to any amino acid selected from the group consisting of nonpolar hydrophobic amino acids, polar uncharged amino acids and polar acidic amino acids.
  • nonpolar hydrophobic amino acids refer to catogorisation of amino acids used by the person skilled in the art.
  • polar uncharged amino acids refer to catogorisation of amino acids used by the person skilled in the art.
  • and polar acidic amio acids refer to catogorisation of amino acids used by the person skilled in the art.
  • non-cationic amino acid comprises the following amino acids: Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
  • oral bioavailability is herein meant the fraction of the administered dose of drug that reaches the systemic circulation after having been administered orally. By definition, when a medication is administered intravenously, its bioavailability is 100%.
  • an insulin peptide may be measured in an assay as known by a person skilled in the art as e.g. described in WO 2005012347.
  • surfactant refers to any substance, in particular a detergent, that can adsorb at surfaces and interfaces, such as but not limited to liquid to air, liquid to liquid, liquid to container or liquid to any solid and which has no charged groups in its hydrophilic groups.
  • permeation enhancer when used herein refers to biologicals or chemicals that promote the absorption of drugs.
  • preservative refers to a chemical compound which is added to a pharmaceutical composition to prevent or delay microbial activity (growth and metabolism).
  • examples of pharmaceutically acceptable preservatives are phenol, m-cresol and a mixture of phenol and m-cresol.
  • macromolecule refers to non-polymeric molecules and comprises nucleic acids, peptides, proteins, carbohydrates, and lipids.
  • polypeptide and “peptide” as used herein means a compound composed of at least two constituent amino acids connected by peptide bonds.
  • the constituent amino acids may be from the group of the amino acids encoded by the genetic code and they may be natural amino acids which are not encoded by the genetic code, as well as synthetic amino acids.
  • natural amino acids which are not encoded by the genetic code are e.g., ⁇ -carboxyglutamate, ornithine, phosphoserine, D-alanine and D-glutamine.
  • synthetic amino acids comprise amino acids manufactured by chemical synthesis, i.e.
  • D-isomers of the amino acids encoded by the genetic code such as D-alanine and D-leucine, Aib (a-aminoisobutyric acid), Abu (a-aminobutyric acid), Tie (tert-butylglycine), ⁇ -alanine, 3-aminomethyl benzoic acid, anthranilic acid.
  • Protein as used herein means a biochemical compound consisting of one or more polypeptides.
  • macromolecule refers to nucleic acids, peptides, proteins, carbohydrates, and lipids as well as non-polymeric molecules with large molecular mass used in therapy and includes without being limited therto insulin, insulin analouges and insulin derivatives.
  • lage molecular mass means a molecular mass above 1500Da.
  • large molecular mass means a molecular mass between 150Da and 6000Da.
  • drug refers to an active ingredient used in a pharmaceutical composition, which may be used in therapy and thus also refer to what was defined as “macromolecular therapeutic” or “therapeutic macromolecule” in the present patent application.
  • insulin peptide an insulin peptide or “the insulin peptide” as used herein is meant human insulin comprising disulfide bridges between CysA7 and CysB7 and between CysA20 and CysB19 and an internal disulfide bridge between CysA6 and CysA1 1 or an insulin analogue or derivative thereof.
  • peptide as used herein comprises also peptides, proteins, conjugates of such peptides and proteins and biologically active fragments thereof.
  • protein comprises peptides and also refers to proteins and biologically active fragments thereof.
  • Human insulin consists of two polypeptide chains, the A and B chains which contain 21 and 30 amino acid residues, respectively. The A and B chains are interconnected by two disulphide bridges. Insulin from most other species is similar, but may contain amino acid substitutions in some positions.
  • insulin as used herein is, if not specified further, an insulin selected from the group consisting of human insulin, insulin analogues and insulin derivatives.
  • An insulin analogue as used herein is a polypeptide, such as a insulin peptide which has a molecular structure which formally may be derived from the structure of a naturally occurring insulin, for example that of human insulin, by deleting and/or substituting at least one amino acid residue occurring in the natural insulin and/or by adding at least one amino acid residue.
  • insulin analogue means a modified insulin wherein one or more amino acid residues of the insulin have been substituted by other amino acid residues and/or wherein one or more amino acid residues have been deleted from the insulin and/or wherein one or more amino acid residues have been added and/or inserted to the insulin.
  • an insulin analogue according to the invention comprises less than 8 modifications (substitutions, deletions, additions) relative to human insulin.
  • an insulin analogue comprises less than 7 modifications (substitutions, deletions, additions) relative to human insulin. In one embodiment an insulin analogue comprises less than 6 modifications (substitutions, deletions, additions) relative to human insulin.
  • an insulin analogue comprises less than 5 modifications (substitutions, deletions, additions) relative to human insulin. In one embodiment an insulin analogue comprises less than 4 modifications (substitutions, deletions, additions) relative to human insulin. In one embodiment an insulin analogue comprises less than 3 modifications (substitutions, deletions, additions) relative to human insulin. In one embodiment an insulin analogue comprises less than 2 modifications (substitutions, deletions, additions) relative to human insulin.
  • insulin derivative refers to chemically modified parent insulin or an analogue thereof, wherein the modification(s) are in the form of attachment of amides, carbohydrates, alkyl groups, acyl groups, esters, PEGylations, and the like.
  • An insulin derivative according to the invention is a naturally occurring insulin or an insulin analogue which has been chemically modified, e.g. by introducing a side chain in one or more positions of the insulin backbone or by oxidizing or reducing groups of the amino acid residues in the insulin or by converting a free carboxylic group to an ester group or to an amide group.
  • Other derivatives are obtained by acylating a free amino group or a hydroxy group, such as in the B29 position of human insulin or desB30 human insulin.
  • acylated insulin covers modification of insulin by attachment of one or more lipophilic substituents optionally via a linker to the insulin peptide.
  • An insulin derivative is thus human insulin, an insulin analogue or insulin peptide which comprises at least one covalent modification such as a side-chain attached to one or more amino acids of the insulin peptide.
  • the naming of the insulin peptide is done according to the following principles: The names are given as mutations and modifications (acylations) relative to human insulin. For the naming of the acyl moiety, the naming is done according to lUPAC nomenclature and in other cases as peptide nomenclature. For example, naming the acyl moiety:
  • octadecanedioyl-Y-L-Glu-OEG-OEG or “17-carboxyheptadecanoyl-Y-L-Glu- OEG-OEG", wherein OEG is short hand notation for the amino acid -
  • the acyl moiety of the modified peptides or proteins may be in the form of a pure enantiomer wherein the stereo configuration of the chiral amino acid moiety is either D or L (or if using the R/S terminology: either R or S) or it may be in the form of a mixture of enantiomers (D and L / R and S).
  • the acyl moiety is in the form of a mixture of enantiomers.
  • the acyl moiety is in the form of a pure enantiomer.
  • the chiral amino acid moiety of the acyl moiety is in the L form.
  • the chiral amino acid moiety of the acyl moiety is in the D form.
  • an insulin derivative in an oral pharmaceutical composition according to the invention is an insulin peptide that is acylated in one or more amino acids of the insulin peptide.
  • an insulin derivative in an oral pharmaceutical composition according to the invention is an insulin peptide that is stabilized towards proteolytic degradation (by specific mutations) and further acylated at the B29-lysine.
  • a non-limiting example of insulin peptides that are stabilized towards proteolytic degradation (by specific mutations) may e.g. be found in WO 2008034881 , which is hereby incorporated by reference.
  • acylated insulin peptides suitable for this invention may be mono-substituted having only one acylation group attached to a lysine amino acid residue in the protease stabilized insulin molecule.
  • acylated insulin peptides suitable for the liquid oral
  • composition of the invention may e.g. be found in WO 2009/1 15469 such as in the passage beginning on page 24 thereof and continuing the next 6 pages.
  • the acylated insulin peptide is selected from the group consisting of:
  • the insulin derivative is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • acylated insulin peptides suitable for the liquid oral
  • composition of the invention may e.g. be found in the PCT application WO201 1068019 such as outlined and exemplified in but not limited to the passage beginning on page 20 line 20 and continuing the next 6 pages, to be published in April 2013.
  • the acylated insulin peptide is selected from the group consisting of N-terminally modified insulin consisting of:
  • A1 (Af.Af-Dimethyl), A14E, Bl tAf.Af-dimethyl), B25H, B29K(/V3 ⁇ 4ctadecanedioyl- gGlu-2xOEG), desB30 human insulin
  • AI CAT.AT-Diethyl AI CAT.AT-Diethyl
  • A14E B1 (AT.AT-diethyl), B25H, B29K(/ ⁇ fOctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 W.W-Dimethyl
  • A14E B1 (/V ⁇ / ⁇ f-dimethyl)
  • B16H B25H
  • A1 (A/ ⁇ Af-Dimethyl), A14E, BI ⁇ AT-dimethyl), B25H, desB27,
  • A1 (Af.Af-Dimethyl), A14E, BI ⁇ /NT-dimethyl), desB27, B29K(Afoctadecanedioyl- gGlu-2xOEG), desB30 human insulin
  • AI G Af-Dimethyl
  • A14E B1 F ⁇ AF-dimethyl
  • B25H desB27
  • A1 (Af./NT-Dimethyl), A14E, B1 (Af.AT-dimethyl), desB27, B29K(Af octadecanedioyl- gGlu), desB30 human insulin
  • AI AT-Dimethyl
  • A14E BI ⁇ .AT-dimethyl
  • B25H B29K(Afoctadecanedioyl- gGlu)
  • desB30 human insulin
  • A1 (AfCarbamoyl), A14E, Bl i/NTCarbamoyl), B25H, B29K(/ ⁇ foctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (/ ⁇ Carbamoyl), A14E, B1 (/ ⁇ Carbamoyl), B25H, B29K(/ ⁇ fhexadecanedioyl-gGlu), desB30 human insulin
  • A1 (ATCarbamoyl), A14E, Bl ⁇ Carbamoyl), B25H, B29K(Afeicosanedioyl-gGlu), desB30 human insulin
  • A1 (AfCarbamoyl), A14E, B1 (AfCarbamoyl), B25H, B29K(Afeicosanedioyl-gGlu-
  • A1 (/ ⁇ Carbamoyl), A14E, B1 (/ ⁇ Carbamoyl), B16H, B25H, B29K(/ ⁇ feicosanedioyl- gGlu-2xOEG), desB30 human insulin
  • A1 (N a Carbamoyl), A14E, B1 (N a Carbamoyl), B25H, desB27,
  • A1 (N a Carbamoyl), A14E, B1 (N a Carbamoyl), B25H, desB27,
  • B29K(N E octadecandioyl-gGlu-2xOEG), desB30 human insulin A1 G(N(alp a)carbamoyl), A14E, B1 F(N(alpha)carbamoyl), desB27, B29K(N(eps)hexadecanedioyl-gGlu), desB30 human insulin
  • AI GCAfcarbamoyl A14E, B1 FtAfcarbamoyl), B16H, desB27, B29K(Neps)- eicosanedioyl-gGlu-2xOEG), desB30 human insulin
  • A1 (/ ⁇ Carbamoyl), A14E, B1 (/ ⁇ Carbamoyl), desB27, B29K(A/3 ⁇ 4ctadecanedioyl- gGlu), desB30 human insulin
  • A1 (AfCarbamoyl), A14E, B1 ( ⁇ Carbamoyl), B16H, B25H, B29K(Afeicosanedioyl- gGlu), desB30 human insulin
  • A1 (AfCarbamoyl), A14E, Bl tAfCarbamoyl), desB27, B29K(Afoctadecanedioyl- gGlu-2xOEG), desB30 human insulin
  • A1 (/ ⁇ Carbamoyl), A14E, Bl tAfcarbamoyl), B25H, B29K(/ ⁇ foctadecanedioyl-gGlu), desB30 human insulin
  • A1 (AfCarbamoyl), A14E, B1 ( ⁇ Carbamoyl), B16H, B25H, B29K(Afeicosanedioyl- gGlu), desB30 human insulin
  • A1 (AfAcetyl), A14E, BI ⁇ Acetyl), B25H, B29K(/ ⁇ fhexadecanedioyl-gGlu), desB30 human insulin
  • A1 (AfAcetyl), A14E, BI ⁇ Acetyl), B25H, desB27, B29K(Afoctadecanedioyl-gGlu), desB30 human insulin
  • A1 (ATAcetyl), A14E, BI ⁇ Acetyl), B25H, B29K(/ ⁇ foctadecandioyl-gGlu-2xOEG), desB30 human insulin
  • A1 (/VDimethylglycyl), A14E, BI ⁇ Dimethylglycyl), B25H, B29K(/ ⁇ foctadecanedioyl- gGlu-2xOEG), desB30 human insulin
  • A1 (/V a 3-(/V,/V-Dimethylamino)propionyl), A14E, ⁇ ⁇ ⁇ ⁇ , ⁇ - dimethylamino)propionyl), B25H, B29K(/ ⁇ foctadecanedioyl-gGlu-2xOEG), desB30 human insulin
  • AI ⁇ - ⁇ /V-Dimethylamino ⁇ utanoyl
  • A14E ⁇ ⁇ ⁇ ⁇ , ⁇ - dimethylamino)butanoyl
  • B25H B29K(/ ⁇ foctadecanedioyl-gGlu-2xOEG)
  • desB30 human insulin
  • A1 (AfDimethylglycyl), A14E, BI ⁇ Dimethylglycyl), B25H, desB27,
  • AI GCAfacetyl A14E, B1 F(/V a acetyl),B25H, desB27, B29K(/V3 ⁇ 4ctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfAcetyl), A14E, BI ⁇ Acetyl), B25H, B29K(/ ⁇ feicosanedioyl-gGlu), desB30 human insulin
  • A1 (AfAcetyl), A14E, BI ⁇ Acetyl), B25H, B29K(/ ⁇ feicosanedioyl-gGlu-2xOEG), desB30 human insulin
  • A1 (AfAcetyl), A14E, BI ⁇ Acetyl), B16H, B25H, B29K(/ ⁇ feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfAcetyl), A14E, BI ⁇ Acetyl), B16H, B25H, B29K(/ ⁇ feicosanedioyl-gGlu), desB30 human insulin
  • A1 (AfDimethylglycyl), A14E, BI ⁇ Dimethylglycyl), B16H, B25H,
  • A-1 (/VTrimethyl), A14E, B-1 (/VTrimethyl), B25H, B29K(/ ⁇ foctadecanedioyl-gGlu-
  • A1 (AfAcetyl), A14E, BI ⁇ Acetyl), desB27, B29K(/ ⁇ foctadecanedioyl-gGlu), desB30 human insulin
  • A1 (AfAcetyl), A14E, BI ⁇ Acetyl), desB27, B29K(/ ⁇ foctadecanedioyl-gGlu-2xOEG), desB30 human insulin
  • A1 (AfAcetyl), A14E, B'l ihTAcetyl), B25H, B29K(/ ⁇ foctadecanedioyl-gGlu), desB30 human insulin
  • AI GCAfAcetyl A14E, B1 F ⁇ Acetyl), desB27, B29K(/ ⁇ feicosanedioyl-gGlu), desB30 human insulin AI GiAFAcetyl), A14E, B1 F ⁇ Acetyl), desB27, B29K(/ ⁇ feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
  • AI G ⁇ Acetyl A14E, B1 F ⁇ Acetyl), B25H, desB27, B29K(/ ⁇ feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfSuccinyl), A14E, B1 (Afsuccinyl), B25H, desB27, B29K(/ ⁇ foctadecanedioyl- gGlu-2xOEG), desB30 human insulin
  • A1 (AfSuccinyl), A14E, Bl ⁇ succinyl), B25H, B29K(/ ⁇ foctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfSuccinyl), A14E, Bl ⁇ succinyl), desB27, B29K(/V3 ⁇ 4ctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfGlutaryl), A14E, B1 (ATglutaryl), B25H, B29K(/ ⁇ foctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfGlutaryl), A14E, B1 (ATglutaryl), desB27, B29K(/ ⁇ foctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfDiglycolyl), A14E, B1 ( ⁇ diglycolyl), B25H, desB27, ⁇ 29 ⁇ ( ⁇ octadecanedioyl- gGlu-2xOEG), desB30 human insulin
  • A1 (AfGlutaryl), A14E, B1 (ATglutaryl), B25H, desB27, B29K(/V3 ⁇ 4ctadecanedioyl- gGlu-2xOEG), desB30 human insulin
  • A1 (AfSuccinyl), A14E, Bl ⁇ succinyl), desB27, B29K(/V3 ⁇ 4ctadecanedioyl-gGlu), desB30 human insulin
  • A1 (AfSuccinyl), A14E, Bl ⁇ succinyl), B25H, desB27, B29K(/ ⁇ feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfSuccinyl), A14E, Bl ⁇ succinyl), desB27, B29K(/ ⁇ feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
  • A1 (AfSuccinyl), A14E, Bl ⁇ succinyl), B16H, desB27, B29K(/ ⁇ feicosanedioyl-gGlu-
  • A1 (AfSuccinyl), A14E, Bl ⁇ succinyl), B25H, B29K(/ ⁇ feicosanedioyl-gGlu-2xOEG), desB30 human insulin
  • A1 (AfSuccinyl), A14E, Bl ⁇ succinyl), desB27, B29K(/ ⁇ feicosanedioyl-gGlu), desB30 human insulin
  • A1 (AfGlutaryl), A14E, B1 (ATglutaryl), desB27, B29K(/ ⁇ feicosanedioyl-gGlu), desB30 human insulin
  • A1 (AfGlutaryl), A14E, B /Nfglutaryl), desB27, B29K(/ ⁇ feicosanedioyl-gGlu-2xOEG), desB30 human insulin
  • A1 (AfGlutaryl), A14E, B1 (Afglutaryl), B25H, B29K(/ ⁇ feicosanedioyl-gGlu-2xOEG), desB30 human insulin
  • an N-terminally modified insulin according to the invention has a peptide part which is selected from the group consisting of the following insulin peptides (i.e. insulins of the invention without N-terminal modifications and without the "lipophilic substituent" or acyl moiety): A14E, B25H, desB30 human insulin; A14H, B25H, desB30 human insulin; A14E, B1 E, B25H, desB30 human insulin; A14E, B16E, B25H, desB30 human insulin; A14E, B25H, B28D, desB30 human insulin; A14E, B25H, B27E, desB30 human insulin; A14E, B1 E, B25H, B27E, desB30 human insulin; A14E, B1 E, B16E, B25H, B27E, desB30 human insulin; A8H, A14E, B25H, desB30 human insulin; A8H, A14E, B25H, desB30 human insulin; A8
  • a N-terminally modified insulin according to the invention has a peptide part which is selected from the group consisting of: A14E, B25H, desB30 human insulin; A14E, B16H, B25H, desB30 human insulin; A14E, B16E, B25H, desB30 human insulin; A14E, desB27, desB30 human insulin; A14E, B16H, desB27, desB30 human insulin; A14E, B25H, B26G, B27G, B28G, desB30 human insulin; B25H, desB30 human insulin and A14E, B25H, desB27, desB30 human insulin.
  • a N-terminally modified insulin according to the invention has a peptide part which is selected from any one of the insulins mentioned above that, in addition, are containing the desB27 mutation.
  • a N-terminally modified insulin according to the invention has a peptide part which is selected from the group consisting of: A14E, B25H, desB27, desB30 human insulin; A14E, B16H, B25H, desB27, desB30 human insulin; A14E, desB27, desB30 human insulin; A14E, B16E, B25H, desB27, desB30 human insulin; and B25H, desB27, desB30 human insulin.
  • a N-terminally modified insulin according to the invention has a peptide part which is selected from any of the above mentioned insulins and, in addition, comprise one or two of the following mutations in position A21 and/or B3 to improve chemical stability: A21 G, desA21 , B3Q, or B3G.
  • a N-terminally modified insulin according to the invention has a peptide part which is selected from the group consisting of: A14E, A21 G, B25H, desB30 human insulin; A14E, A21 G, B16H, B25H, desB30 human insulin; A14E, A21 G, B16E, B25H, desB30 human insulin; A14E, A21 G, B25H, desB27, desB30 human insulin; A14E, A21 G, B25H, desB27, desB30 human insulin; A14E, A21 G, B25H, B26G, B27G, B28G, desB30 human insulin; A21 G, B25H, desB30 human insulin and A21 G, B25N, desB30 human insulin, and, preferably, it is selected from the following protease stabilised insulins: A14E, A21 G, B25H, desB30 human insulin; A14E, A21 G, desB27, desB30 human insulin;
  • acylated insulin covers modification of insulin by attachment of one or more lipophilic substituents optionally via a linker to the insulin peptide.
  • a “lipophilic substituent” is herein understood as a side chain consisting of a fatty acid or a fatty diacid attached to the insulin, optionally via a linker, in an amino acid position such as LysB29, or equivalent.
  • the insulin peptide may be present in an amount of a pharmaceutical composition according to the invention in up to about 20% such as up to about 10% by weight of the total pharmaceutical composition, or from about 0.1 % such as from about 1 %. In one embodiment of the invention, the insulin peptide is present in an amount from about 0.1 % to about 20%, in a further embodiment from about 0.1 % to 15%, 0.1 % to 10%, 1 % to 8% or from about 1 % to 5% by weight of the total composition. It is intended, however, that the choice of a particular level of insulin peptide will be made in accordance with factors well-known in the
  • solubility of the insulin peptide in the polar organic solvent or optional hydrophilic component or surfactant used, or a mixture thereof mode of administration and the size and condition of the patient.
  • Each unit dosage will suitably contain from 1 mg to 200 mg insulin peptide, e.g. about 1 mg, 5 mg, 10 mg, 15 mg, 25 mg, 50 mg, 80 mg, 90 mg, 100 mg, 150 mg, 200 mg insulin peptide, e.g. between 5 mg and 200 mg of insulin peptide.
  • each unit dosage contains between 10 mg and 200 mg of insulin peptide.
  • a unit dosage form contains between 10 mg and 100 mg of insulin peptide.
  • the unit dosage form contains between 20 mg and 80 mg of insulin peptide. In yet a further embodiment of the invention, the unit dosage form contains between 30 mg and 60 mg of insulin peptide.
  • the unit dosage form contains between 30 mg and 50 mg of insulin peptide.
  • Such unit dosage forms are suitable for administration 1 -5 times daily depending upon the particular purpose of therapy.
  • polypeptides and peptides such as insulin is well known in the art.
  • Polypeptides or peptides may for instance be produced by classical peptide synthesis, e.g. solid phase peptide synthesis using t-Boc or Fmoc chemistry or other well established techniques, see e.g. Greene and Wuts, "Protective Groups in Organic Synthesis", John Wiley & Sons, 1999.
  • the polypeptides or peptides may also be produced by a method which comprises culturing a host cell containing a DNA sequence encoding the (poly)peptide and capable of expressing the (poly)peptide in a suitable nutrient medium under conditions permitting the expression of the peptide.
  • the recombinant cell should be modified such that the non-natural amino acids are incorporated into the (poly)peptide, for instance by use of tRNA mutants.
  • microemulsion preconcentrate means a composition, which spontaneously forms a microemulsion or a nanoemulsion, e.g., an oil-in-water microemulsion or nanoemulsion, swollen micelle, micellar solution, in an aqueous medium, e.g. in water or in the gastrointestinal fluids after oral application.
  • the composition self- emulsifies upon dilution in an aqueous medium for example in a dilution of 1 :5, 1 :10, 1 :50, 1 :100 or higher.
  • the composition according to the present invention forms the microemuslion or nanoemulsion comprising particles or domains of a size below 100nm in diameter.
  • domain size refers to repetitive scattering units and may be measured by e.g., small angle X-ray.
  • the domain size is smaller than 150nm, in another embodiment, smaller than 100nm and in another embodiment, smaller than 50nm, in another embodiment, smaller than 20nm, in another embodiment, smaller than 15nm, in yet another embodiment, smaller than 10nm.
  • SEDDS self emulsifying drug delivery systems
  • a hydrophilic component a surfactant, optionally a co-surfactant or lipid component and a therapeutic macromolecule that forms spontaneously a fine oil in water emulsion when exposed to aqueous media under conditions of gentle agitation or digestive motility that would be encountered in the Gl tract.
  • SMEDDS self micro-emulsifying drug delivery systems
  • SNEDDS self nano-emulsifying drug delivery systems
  • emulsion refers to a slightly opaque, opalescent or opague colloidal coarse dispersion that is formed spontaneously or substantially
  • microemulsion refers to a clear or translucent, slightly opaque, opalescent, non-opaque or substantially non-opaque colloidal dispersion that is formed spontaneously or substantially spontaneously when its components are brought into contact with an aqueous medium.
  • a microemulsion is thermodynamically stable and contains homogenously dispersed particles or domains, for example of a solid or liquid state (e.g., liquid lipid particles or droplets), of a mean diameter of less than 150 nm as measured by standard light scattering techniques, e.g., using a MALVERN ZETASIZER Nano ZS.
  • a microemulsion is formed which contains homogenously dispersed particles or domains of a mean diameter of less than 100nm, such as less than 50nm, less than 40nm and less than 30nm.
  • Z average (nm) refers to the partice size of the particles or domains of said microemulsion.
  • PDI polydispersity index
  • domain size refers to repetitive scattering units and may be measured by e.g., small angle X-ray. In one embodiment of the invention, the domain size is smaller than 150 nm, In one embodiment, smaller than 100 nm and In one embodiment, smaller than 50 nm, In one embodiment, smaller than 20 nm, In one embodiment, smaller than 15 nm, in yet another embodiment, smaller than 10 nm.
  • nanoemulsion refers to a clear or translucent, slightly opaque, opalescent, non-opaque or substantially non-opaque colloidal dispersion with particle or droplet size below 20 nm in diameter (as e.g. measured by PCS) that is formed spontaneously or substantially spontaneously when its components are brought into contact with an aqueous medium.
  • a microemulsion is formed which contains homogenously dispersed particles or domains of a mean diameter of less than 20 nm, such as less than 15 nm, less than 10 nm and greater than about 2-4 nm.
  • spontaneously dispersible when referring to a pre- concentrate refers to a composition that is capable of producing colloidal structures such as nanoemulsions, microemulsions, emulsions and other colloidal systems, when diluted with an aqueous medium when the components of the composition of the invention are brought into contact with an aqueous medium, e.g. by simple shaking by hand for a short period of time, for example for ten seconds.
  • a spontaneously dispersible concentrate according to the invention is a SEDDS, SMEDDS or SNEDDS.
  • non-ionic surfactant refers to any substance, in particular a detergent, that can adsorb at surfaces and interfaces, like liquid to air, liquid to liquid, liquid to container or liquid to any solid and which has no charged groups in its hydrophilic group(s) (sometimes referred to as "heads").
  • the non-ionic surfactant may be selected from a detergent such as ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglyceri- des and sorbitan fatty acid esters, polysorbate such as polysorbate-20, polysorbate-40, polysorbate-60, polysorbate-80, super refined polysorbate 20, super refined polysorbate 40, super refined polysorbate 60 and super refined polysorbate 80 (where the term "super refined” is used by the supplier Croda for their high purity Tween products), poloxamers such as poloxamer 188 and poloxamer 407, polyoxyethylene sorbitan fatty acid esters,
  • a detergent such as ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglyceri- des and sorbitan fatty acid esters
  • polysorbate such as polysorbate-20, polysorbate-40, polysorbate-60, polysorbate-80
  • polyoxyethylene derivatives such as alkylated and alkoxylated derivatives (Tweens, e.g. Tween-20 or Tween-80), block copolymers such as polyethyleneoxide/polypropyleneoxide block copolymers (e.g. Pluronics/Tetronics, Triton X-100 and/or Synperonic PE/L 44 PEL) and ethoxylated sorbitan alkanoates surfactants (e. g.
  • Tween-20, Tween-40, Tween-80, Brij- 35 diglycerol laurate, diglycerol caprate, diglycerol caprylate, diglycerol monocaprylate, polyglycerol laurate, polyglycerol caprate and polyglycerol caprylate.
  • non-aqueous refers to a composition to which no water is added during preparation of the pharmaceutical composition. It is known to the person skilled in the art that a composition which has been prepared without addition of water may take up small amounts of water from the surroundings during handling of the pharmaceutical composition such as e.g. a soft-capsule or a hard-capsule used to encapsulate the composition. Also, the insulin peptide and/or one or more of the excipients in the
  • a non-aqueous pharmaceutical composition according to the invention may thus contain small amounts of water.
  • a non-aqueous pharmaceutical composition according to the invention comprises less than 10%(w/w) water.
  • the composition according to the invention comprises less than 5%(w/w) water.
  • the composition according to the invention comprises less than 4%(w/w) water, in another embodiment less than 3%(w/w) water, in another embodiment less than 2%(w/w) water and in yet another embodiment less than 1 %(w/w) water .
  • the composition accord 0%(w/w) water
  • non-ionic surfactants examples include, but are not limited to:
  • reaction products of a natural or hydrogenated castor oil and ethylene oxide may be reacted with ethylene oxide in a molar ratio of from about 1 :35 to about 1 :60, with optional removal of the PEG component from the products.
  • Various such surfactants are commercially available, e.g., the CREMOPHOR series from BASF Corp. (Mt.
  • CREMOPHOR RH 40 which is PEG40 hydrogenated castor oil which has a saponification value of about 50- to 60, an acid value less than about one, a water content, i.e., Fischer, less than about 2%, an n D 60 of about 1.453-1 .457, and an HLB of about 14-16; 2.
  • Polyoxyethylene fatty acid esters that include polyoxyethylene stearic acid esters, such as the MYRJ series from Uniqema e.g., MYRJ 53 having a m.p. of about 47°C.
  • MYRJ 53 having an m.p. of about 47°C and PEG-40-stearate available as MYRJ 52;
  • Sorbitan derivatives that include the TWEEN series from Uniqema, e.g., TWEEN 60;
  • Polyoxyethylene-polyoxypropylene co-polymers and block co-polymers or poloxamers e.g., Pluronic F127 or Pluronic F68 from BASF or Synperonic PE/L from Croda;.
  • Polyoxyethylene alkyl ethers e.g., such as polyoxyethylene glycol ethers of Ci 2 -Ci 8 alcohols, e.g., polyoxyl 10- or 20-cetyl ether or polyoxyl 23-lauryl ether, or 20-oleyl ether, or polyoxyl 10-, 20- or 100-stearyl ether, as known and commercially available as the BRIJ series from Uniqema.
  • Particularly useful products from the BRIJ series are BRIJ 58; BRIJ 76; BRIJ 78; BRIJ 35, i.e. polyoxyl 23 lauryl ether; and BRIJ 98, i.e., polyoxyl 20 oleyl ether.
  • These products have a m.p. between about 32°C to about 43°C;
  • PEG sterol ethers having, e.g., from 5-35 [CH 2 -CH,-0] units, e.g., 20-30 units, e-g., SOLULAN C24 (Choleth-24 and Cetheth-24) from Chemron (Paso Robles, CA); similar products which may also be used are those which are known and commercially available as NIKKOL BPS-30 (polyethoxylated 30 phytosterol) and NIKKOL BPSH-25 (polyethoxylated 25 phytostanol) from Nikko Chemicals;
  • Polyglycerol fatty acid esters e.g., having a range of glycerol units from 4-10, or 4, 6 or 10 glycerol units.
  • particularly suitable are deca-/hexa-/tetraglyceryl monostearate, e.g., DECAGLYN, HEXAGLYN and TETRAGLYN from Nikko Chemicals;
  • Alkylene polyol ether or ester e.g., lauroyl macrogol-32 glycerides and/or stearoyl macrogol-32 glycerides which are GELUCIRE 44/14 and GELUCIRE 50/13 respectively;
  • hydroxy fatty acid e.g. 12 hydroxy stearic acid PEG ester, e.g. of PEG about e.g. 600-900 e.g. 660 Daltons MW, e.g. SOLUTOL HS 15 from BASF (Ludwigshafen, 20 Germany).
  • SOLUTOL HS 15 comprises about 70% polyethoxylated 12-hydroxystearate by weight and about 30% by weight unesterified polyethylene glycol component. It has a hydrogenation value of 90 to 1 10, a saponification value of 53 to 63, an acid number of maximum 1 , and a maximum water content of 0.5% by weight; 1 1.
  • Polyoxyethylene-polyoxypropylene-alkyl ethers e.g. polyoxyethylene-polyoxypropylene- ethers of Q 2 to C
  • Polyethoxylated distearates e.g. commercially available under the tradenames ATLAS G 1821 from Uniqema and NIKKOCDS-6000P from Nikko Chemicals.
  • HLB Hydrophilic-lipophilic balance
  • a surfactant or lipophilic component is a measure of the degree to which it is hydrophilic or lipophilic, determined by calculating values for the different regions of the molecule, as described by Griffin (Griffin WC: “Classification of Surface-Active Agents by 'HLB,'” Journal of the Society of Cosmetic Chemists 1 (1949): 31 1 ) or by Davies (Davies JT: "A quantitative kinetic theory of emulsion type, I. Physical chemistry of the emulsifying agent," Gas/Liquid and
  • Non-ionic surfactants with HLB above 10 are a selection of non-ionic surfactants which have the common feature of having HLB above 10.
  • Diglycerol caprylate diglycerol monocaprylate, polyglycerol caprylate
  • HLB 1 1
  • Polyglycerol caprate (Rylo PG10 Pharma) with HLB of 10;
  • Caprylocaproyl macrogolglycerides (Labrasol, Labrasol ALF) with an HLB of 14;
  • Block polymers such as SYNPERONIC PE/L 44 (Poloxamer 124);
  • Polyoxyethylenestearate (Myrj 45, Macrogolstearate) with HLB of 1 1 .1 ;
  • Polyoxyethylenestearate (Myrj 49, Macrogolstearate) with HLB of 15;
  • amino acid refers to any molecule that contains both amine and carboxyl functional groups.
  • enteric coating means a polymer coating that controls disintegration and release of the solid oral dosage form.
  • the site of disintegration and release of the solid dosage form may be designed depending on the pH of the targeted area, where absorbtion of the therapeutic macromolecule (i.e. therapeutical active peptide or protein) is desired, thus does also include acid resistant protective coatings.
  • enteric coatings includes known enteric coatings, but also any other coating with enteric properties, wherein said term “enteric properties” means properties controlling the disintegration and release of the solid oral dosage form (i.e. the oral pharmaceutical composition according to this invention).
  • enteric soft- or hard capsule technology when used herein means soft- or hard capsule technology comprising at least one element with enteric properties, such as at least one layer of an enteric coating.
  • delayed release coatings as used herein means a polymer coating which releases the API in a delayed manner after oral dosing. Delayed release can be achieved by pH dependent or pH independent polymer coatings.
  • co-surfactant when used herein refers to an additional surfactant added to a composition or formulation, wherein a first surfactant is present.
  • An oral pharmaceutical composition comprising
  • R1 is a fatty acid chain comprising 8 to 18 carbon atoms
  • R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group)
  • R3 is either H, or a salt of
  • R4 is a non-cationic amino acid side chain and b. at least one therapeutic macromolecule.
  • An oral pharmaceutical composition comprising
  • R1 is a fatty acid chain comprising 8 to 18 carbon atoms
  • R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), and
  • R3 is either H, or a salt thereof
  • R4 is a non-cationic amino acid side chain
  • An oral pharmaceutical composition comprising
  • R1 is a fatty acid chain comprising 8 to 18 carbon atoms
  • R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), and
  • R3 is either H, or a salt thereof
  • R4 is a non-cationic amino acid side chain
  • composition according to any of the preceding aspects, wherein the amino acid residue of said at least one fatty acid acylated amino acid is based on a nonpolar hydrophobic amino acid.
  • an oral pharmaceutical composition according to any of the preceding aspects wherein the amino acid residue of said at least one fatty acid acylated amino acid is based on a polar uncharged amino acid.
  • An oral pharmaceutical composition according to any of the preceding aspects wherein the amino acid residue of said at least one fatty acid acylated amino acid is a based on a polar acidic amino acid.
  • a solid oral composition according to any of the preceeding aspects further comprising at least one insulin.
  • a solid oral composition according to any of the preceedingaspects further comprising an enteric or delayed release coating.
  • amino acid residue of said FA-aa is selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and
  • Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
  • amino acid residue of said FA-aa is selected from the group consisting of the form of the free acid or salt of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
  • the FA-aa is selected from the group consisting of: Sodium lauroyl alaninate, N-dodecanoyl-L-alanine, Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium lauroyi glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyi glycinate, N-dodecanoyl-L-glycine, Sodium lauroyi histidinate, N-dodecanoyl-L- histidine, Sodium
  • N-decanoyl-L-phenylalanine Sodium capric prolinate, N-decanoyl-L- proline, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N- decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L-tryptophane,
  • Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyi sarcosinate, Sodium N- decyl leucine, Sodium cocoyl glycine and Sodium cocoyl glutamate.
  • compositions according to any of the preceeding aspects, wherein the FA-aa is selected from the group consisting of: Sodium lauroyi alaninate, N-dodecanoyl-L-alanine, Sodium lauroyi asparaginate, N-dodecanoyl-L-asparagine,
  • L-tyrosine Sodium lauroyi valinate, N-dodecanoyl-L-valine, Sodium lauroyi sarcosinate, N-dodecanoyl-L-sarcosine, Sodium capric alaninate, N-decanoyl-L- alanine, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N-decanoyl-L- cysteine, Sodium capric glutamic acid, N-decanoyl-L-glutamic acid, Sodium capric glutaminate, N-decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric histidinate, N-decanoyl-L-histidine, Sodium
  • Amisoft MS-1 1 (Sodium Myristoyl Glutamate)
  • Amisoft LS-1 1 (Sodium Lauroyi Glutamate)
  • Amisoft CS-1 1 (Sodium Cocoyl Glutamate)
  • Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyi sarcosinate, Sodium N-decyl leucine and Sodium cocoyl glycine, Sodium cocoyl glutamate.
  • An oral pharmaceutical composition according to any of the preceeding aspects further comprising propylene glycol.
  • An oral pharmaceutical composition according to any of the preceeding aspects further comprising SEDDS, SMEDDS or SNEDDS.
  • composition according to any of the preceding aspects, which comprises less than 10%(w/w) water.
  • amino acid residue of said at least one fatty acid acylated amino acid is based on a nonpolar hydrophobic amino acid, a polar uncharged amino acid or polar acidic amino acid.
  • composition according to any of the preceeding aspects further comprising an enteric or delayed release coating.
  • amino acid residue of said FA-aa is selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine
  • Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
  • fatty acid acylated amino acid is selected from the group consisting of: Sodium lauroyl alaninate, N-dodecanoyl-L-alanine, Sodium lauroyl asparaginate, N- dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyl glutamic acid, N- dodecanoyl-L-glutamic acid, Sodium lauroyl glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyl glycinate, N-dodecanoyl-L-glycine, Sodium lauroyl histidinate, N- dodecanoyl-L-hist
  • N-decanoyl-L-proline Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L- tryptophane, Sodium capric tyrosinate, N-decanoyl-L-tyrosine, Sodium capric valinate, N-decanoyl-L-valine, Sodium capric sarcosinate and N-decanoyl-L-sarcosine, Sodium lauroyi sarcosinate, Sodium oleoyl sarcosinate, Sodium N-decyl leucine, Amisoft HS-
  • Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyi sarcosinate, Sodium N-decyl leucine, Sodium cocoyl glycine, Sodium cocoyl glutamate Sodium lauroyi alaninate, N-dodecanoyl-L-alanine, Sodium lauroyi asparaginate, N- dodecanoyl-L-asparagine, Sodium lauroyi aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyi cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyi glutamic acid, N- dodecanoyl-L-glutamic acid, Sodium lauroyi glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyi glycinate, N-dodecanoyl-L-glycine
  • Amilite GCS-1 1 Sodium Cocoyl Glycinate
  • Sodium lauroyl sarcosinate Sodium N-decyl leucine
  • Sodium cocoyl glycine Sodium cocoyl glutamate.
  • the oral pharmaceutical composition according to any of the preceeding aspects further comprising propylene glycol.
  • An oral pharmaceutical composition according to any of the preceeding aspects, further comprising SEDDS, SMEDDS or SNEDDS.
  • oral pharmaceutical composition according to any of the preceeding aspects, further comprising other pharmaceutical excipients.
  • the oral pharmaceutical composition according to any of the preceeding aspects for use as a medicament.
  • the oral pharmaceutical composition according to any of the preceeding aspects for use as a medicament for treatment of Diabetes Mellitus.
  • an oral pharmaceutical composition for increasing the bioavailability of said therapeutic active peptide.
  • Examples of pharmaceutical compositions comprising insulin derivative and fatty acid acylated amino acids.
  • the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin 60 nmol/kg dissolved in phosphate buffer (pH 7.4) in presence of fatty acid acylated amino acids.
  • the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin 60 nmol/kg dissolved in phosphate buffer (pH 7.4) in presence of sodium N- capric leucine in concentrations of 10 or 20mg/ml_, respectively.
  • the formulation (- ⁇ -) with N-cocoyl sarcosine contained 50% of the co-solvent propylene glycol.
  • the fatty acid chain distribution in the cocoyl sarcosinate was 1 % C6, 8% C8, 6% C10, 48% C12, 18% C14, 8% C16, 6% C18 saturated and 5% C18 unsaturated.
  • the insulin derivative A14E, B25H, B29K N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of increasing amounts (3mg/ml_, 10mg/ml_, 30mg/ml_ and 100mg/L) of sodium lauroyl sarcosinate
  • the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin 60 nmol/kg dissolved in phosphate buffer (pH 7.4) in presence of increasing amounts (3mg/ml_, 10mg/ml_, 30mg/ml_ and 100mg/L) of sodium myristoyl glutamate.
  • the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin 60 nmol/kg dissolved in phosphate buffer (pH 7.4) in presence of 10 mg/mL sodium lauroyl sarcosinate.
  • the fatty acid chain distribution in the cocoyi sarcosinate is 1 % C6, 8% C8, 6% C10, 48% C12, 18% C14, 8% C16, 6% C18 saturated and 5% C18 unsaturated.
  • the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin 60 nmol/kg dissolved in phosphate buffer (pH 7.4) in presence of 10 mg/mL sodium N-capric leucine, sodium, N-capric alanine, sodium N-capric phenylalanine, N-capric isoleucine, N-capric aspart, N-lauroyl leucine or N-myristoyl leucine, respectively.
  • the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin 60 nmol/kg dissolved in propylene glycol in presence of sodium N-capric leucine.
  • Pharmakokinetic profiles were retrieved from the resulting records related to measurements after peroral dosing of an enteric coated tablet comprising 200 mg of sodium lauroyl sarcosinate, 50 mg of soybean trypsine inhibitor (SBTI) and Eudragit® L30-D55 and Eudragit® NE30D for enteric coating further comprising insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG) and desB30 human insulin (120 nmol/kg) after peroral dosing of an enteric coated tablet comprising 200 mg of sodium lauroyl sarcosinate, 50 mg of soybean trypsine inhibitor (SBTI) and Eudragit® L30-D55 and Eudragit® NE30D for enteric coating to male beagle dogs.
  • SBTI soybean trypsine inhibitor
  • Eudragit® L30-D55 and Eudragit® NE30D for enteric coating to male beagle dogs
  • the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin 60 nmol/kg dissolved in phosphate buffer (pH 7.4) in presence of 10mg sodium lauroyl leucine, a mixtures of 5 mg/mL sodium lauroyl leucine and 5mg/ml_ capric leucine or 10 mg/mL of the commonly used permeation enhancers salicylate, deoxycholate.
  • Liquid insulin SEDDS, SMEDDS and SNEDDS formulations were prepared according to the guidance given in WO08145728 comprising the fatty acid acylated amino acid sodium N- lauroyl phenylalanine.
  • All formulations contained the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl- gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg).
  • compositions are shown in table 1 and PK results are shown in Figure 13.
  • compositions are shown in Table 1.
  • Table 1 Liquid insulin SEDDS, SMEDDS and SNEDDS formulations comprising the co- surfactant sodium N-lauroyl phenylalanine. All formulations contain the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl- gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg).
  • Insulin SEDDS and SMEDDS compositions were prepared according to the guidance given in WO08145728 comprising at least one fatty acid acylated amino acid (FA-aa). Mean particle size (hydrodynamic diameter) was analysed after 100 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index). All formulations contained the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg).
  • Table 2 Insulin SEDDS and SMEDDS compositions comprising at least one FA-aa.
  • Mean particle size (hydrodynamic diameter) is shown after 100 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index).
  • Insulin Propylene FA-aa 50 Tween Diglycerol
  • Insulin Propylene FA-aa 50 Tween Diglycerol
  • Example 15 Enteric softcapsule comprising insulin derivative and fatty acid acylated amino acids formulated in a SEDDS.
  • Insulin SEDDS compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa).
  • Pharmakokinetic profile in a single beagle dog is shown of the insulin derivative A1 (N,N- Dimethyl), A14E, B1 (N, N-dimethyl), B25H, B29K(N(eps)octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (120 nmol/kg) after peroral dosing of an enteric coated soft capsule comprising 30 mg of sodium lauroyi leucine sodium salt, 150 mg of propylene glycol, 300 mg of Polysorbate 20 and 520 mg of diglycerol monocaprylate.
  • Liquid non-aqueous insulin analogue compositions with different amounts of N-lauroyl leucine sodium salt Liquid non-aqueous insulin analogue compositions with different amounts of N-lauroyl leucine sodium salt.Jnsulin SEDDS compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa). Insulin SEDDS and SMEDDS compositions were prepared comprising increasing amounts of N-lauroyl leucine sodium salt.
  • Liquid insulin analogue compositions with different amounts of N-lauroyl leucine sodium salt further comprising diglycerol monocaprylate and propylene glycol.
  • compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa).
  • nsulin SEDDS compositions were prepared comprising different amounts of N-lauroyl leucine sodium salt. Mean particle size
  • All formulations contained the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (5mg/g).
  • Insulin SEDDS compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa).
  • Insulin SEDDS and SMEDDS compositions were prepared comprising different fatty acid acylated aminoacid sodium salts, polysorbates and solvents.
  • Liquid lipid based formulations comprising at least one fatty acid acylated aminoacid, insulin derivative, solvent and at least one lipid or co-surfactant were prepared.
  • Insulin SEDDS compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa).
  • Insulin SEDDS compositions were prepared comprising different fatty acid acylated aminoacid sodium salts, lipid or co-surfactant and a solvent.
  • Mean particle size fatty acid acylated aminoacid sodium salts, lipid or co-surfactant and a solvent.
  • Liquid lipid based formulations comprising at least one fatty acid acylated aminoacid, insulin derivative, solvent and at least one lipid or co-surfactant are described.
  • Lipid SEDDS, SMEDDS and SNEDDS compositions comprising N-lauroyl leucine sodium salt and different surfactants with variable HLB values were prepared.
  • Insulin SEDDS compositions were prepared according to the guidance given in WO08145728 comprising at least one fatty acid acylated amino acid (FA-aa).
  • Insulin SEDDS and SMEDDS compositions were prepared comprising N-lauroyl leucine sodium salt, propylene glycol, diglycerol mono caprylate and a high or low HLB surfactant.
  • All formulations comprise 5 mg/g insulin analogue A1(N,N-Dimethyl), A14E, B1(N, N- dimethyl), B25H, B29K(N(eps)octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin.
  • composition of the insulin degludec/liraglutide drug product that Novo Nordisk A/S currently has in clinical development is shown below. This formulation has been shown to be a stable combination product suitable for use in type II diabetes clinical trials (subcutaneous injection)."
  • Insulin degludec 600 nmol (100 U) per ml
  • Both insulin degludec and liraglutide drug substances are added in the form of a solid powder, separately and directly to a mixture of excipients.

Abstract

The present invention relates to fatty acid acylated amino acids (FA-aa's) acting as permeation enhancers for oral delivery of therapeutic macromolecules such as peptides and pharmaceutical compositions comprising such FA-aa's.

Description

FATTY ACID ACYLATED AMINO ACIDS FOR ORAL PEPTIDE DELIVERY
TECHNICAL FIELD
The technical field of this invention relates to fatty acid acylated amino acids (FA- aa's) for oral delivery of therapeutic hydropilic peptides and proteins and pharmaceutical compositions comprising such FA-aa's.
BACKGROUND
Many pathological states due to deficiencies in or complete failure of the production of a certain macromolecules (e.g. proteins and peptides) are treated with an invasive and inconvenient parenteral administration of therapeutic macromolecules, such as hydrophilic peptides or proteins. One example hereof is the administration of insulin in the treatment of insulin dependent patients, who are in need of one or more daily doses of insulin. The oral route is desirable for administration due to its non-invasive nature and has a great potential to decrease the patient's discomfort related to drug administration and to increase drug compliance. However several barriers exist; such as the enzymatic degradation in the gastrointestinal (Gl) tract, drug efflux pumps, insufficient and variable absorption from the intestinal mucosa, as well as first pass metabolism in the liver and until now no products for oral delivery of therapeutic hydrophilic proteins are found to be marketed.
A non-limiting example of a hydrophilic proteins and polypeptides is human insulin which ais degraded by various digestive enzymes found in the stomach (pepsin), in the intestinal lumen (chymotrypsin, trypsin, elastase, carboxypeptidases, etc.) and in the mucosal surfaces of the Gl tract (aminopeptidases, carboxypeptidases, enteropeptidases, dipeptidyl peptidases, endopeptidases, etc.).
WO2004147578 relates to fatty acid acylated amino acids used as permeation enhancers for hydrophobic molecules including hydrophobic macromolecules such as cyclosporine.
WO2001035998 relates to acylated amino acids used as transdermal and transmucosal absorption promoters for macromolecules, such as hydrophilic peptides or proteins.
WO2004064758 relates to an oral composition for delivering pharmaceutical peptides, such as insulin, growth hormone and GLP-1 , comprising absorption enhancers, including acyl amino acids.
US2005282756 is related to a dry powder composition comprising insulin and an absorption enhancer. WO2003030865 is related to insulin compositions comprising surfactants such as ionic surfactants and does also contain oil or lipid compounds such as triglycerides and does further comprise long chain esterified fatty acids (C12 to C18).
WO2004064758 is related to an oral pharmaceutical composition for delivering pharmaceutical peptides, comprising absorption enhancers.
The oral route of administration is rather complex and a need for establishment of an acceptable composition suitable for the treatment of patients, with an effective
bioavailability of the macromolecule, such as hydrophilic peptides or proteins, is existent.
SUMMARY
This invention is an oral pharmaceutical composition comprising certain amino acids acylated at their alpha-amino group with a fatty acid of 8 to 18 carbons and and an active ingredient, such as a hydrophilic peptide or protein.
BRIEF DESCRIPTION OF DRAWINGS
Figure 1. Pharmakokinetic profiles of the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of a fatty acid acylated amino after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 6)
Figure 2. Pharmakokinetic profiles of the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of sodium N-capric leucine in two different concentrations after injection into mid-jejunum of anaesthetized overnight fasted Sprague- Dawley rats (n = 4-6). Figure 3. Pharmakokinetic profiles of the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of two different fatty acid acylated amino acids after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 6). The formulation (-□-) with N-cocoyl sarcosine contained 50% of the co-solvent propylene glycol. The fatty acid chain distribution in the cocoyl sarcosinate is 1 % C6, 8% C8, 6% C10, 48% C12, 18% C14, 8% C16, 6% C18 saturated and 5% C18 unsaturated. Figure 4. Pharmakokinetic profiles of the insulin derivative A14E, B25H, B29K (N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of increasing amounts of sodium lauroyi sarcosinate after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 6).
Figure 5. Pharmakokinetic profiles of the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of increasing amounts of sodium myristoyl glutamate after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 4-6).
Figure 6. Pharmakokinetic profiles of the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of 10 mg/ml sodium lauroyi sarcosinate after injection into the colon of anaesthetized overnight fasted Sprague-Dawley rats.
Figure 7. Pharmakokinetic profiles of the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of oleoyi sarcosinate or in presence of cocoyl sarcosinate and 16.5% of the co-solvent propylene glycol after injection into mid- jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 6). The fatty acid chain distribution in the cocoyl sarcosinate is 1 % C6, 8% C8, 6% C10, 48% C12, 18% C14, 8% C16, 6% C18 saturated and 5% C18 unsaturated.
Figure 8. Pharmakokinetic profiles of the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of different fatty acid acylated amino acids after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 4-6).
Figure 9. Pharmakokinetic profiles of the insulin derivative A14E, B25H,
B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of different fatty acid acylated amino acids after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 5-6).
Figure 10. Pharmakokinetic profiles of the insulin derivative A14E, B25H,
B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in propylene glycol in presence of sodium N-capric leucine after injection into mid- jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 6).
Figure 11. Pharmakokinetic profiles of the insulin derivative A14E, B25H,
B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (120 nmol/kg) after peroral dosing of an enteric coated tablet comprising 200 mg of sodium lauroyl sarcosinate and 50 mg of soybean trypsine inhibitor and Eudragit® L30D55 and Eudragit® NE30D for enteric coating to male beagle dogs. Figure 12. Pharmakokinetic profiles of the insulin derivative A14E, B25H,
B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of different fatty acid acylated amino acids or mixtures thereof according to the invention or in presence of commonly used permeation enhancers after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 5-6).
Figure 13. Pharmakokinetic profiles of the insulin derivative A14E, B25H,
B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg) dissolved in liquid SEDDS, SMEDDS and SNEDDS formulations comprising sodium N- lauroyl phenylalanine after injection into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 5-6). The compositions are shown in table 1 .
Figure 14. Pharmakokinetic profile in a single beagle dog is shown of the insulin derivative A1 (N,N-Dimethyl), A14E, B1 (N, N-dimethyl), B25H, B29K(N(eps)octadecanedioyl-gGlu- OEG-OEG), desB30 human insulin (120 nmol/kg) after peroral dosing of an enteric coated soft capsule comprising 30mg of sodium lauroyl leucine sodium salt, 150mg of propylene glycol, 300mg of Polysorbate 20 and 520mg of diglycerol monocaprylate.
A 1 :1 mixture of Euragit® L30-D55 and Eudragit® NE30D was used for enteric coating. DESCRIPTION
The present invention is related to pharmaceutical compositions, comprising FA-aa's acting as permeation enhancers suitable for oral administration of therapeutic
macromolecules (e.i. therapeutic acvtive peptides and proteins). More specifically therapeutic macromolecules, such as hydrophilic peptides or proteins according to the present invention are hydrophilic peptides and proteins which have a therapeutical activity and include but are not limited to insulin. The research into new surfactants with low irritant action has lead to the development of different surfactants derived from amino acids (Mitjans et al., 2003;
Benavides et al., 2004; Sanchez et al., 2006) FA-aa's are amino acid based surfactants and thus mild biodegradable surfactants with a low toxicity.
It has surprisingly been found that certain fatty acid N-acylated amino acids increase the absorption of hydrophillic peptides and proteins after oral administration to a higher degree than commonly used permeation enhancers known in the art such as fatty acid salts, bile salts and others. This effect has been shown for hydrophilic peptides and proteins of varying sizes.
Due to their low toxicity and increasing effect on oral bioavailability of the therapeutic macromolecule, such as a hydrophilic peptide or protein, FA-aa's according to the present invention are valuable ingredients in oral pharmaceutical compositions. Especially valuable are FA-aa's according to this invention in oral pharmaceutical compositions comprising hydrophilic peptides or proteins as active ingredient.This is of interest for diseases that demand chronic administration of therapeutic macromolecules (e.g. peptides or proteins), but is not limited hereto, since the most non-invasive, non-toxic administration of drugs is generally favoured in any treatment, also for sporadic or bulk administration of therapeutics. So far, there are no commercial hydrophilic proteins available as oral formulation mainly due to the great challenges of enzymatic degradation and very low intestinal permeability of such hydrophilic proteins and peptides. Foger et al. described the impact of the molecular weight on oral absorption of hydrophilic peptide drugs and showed that the permeability decreased with increasing molecular weight of such hydrophilic peptide drugs (Amino Acids (2008) 25: 233-241 , DOI 10.1007/s00726-007-0581 -5).
The invention may also solve further problems that will be apparent from the disclosure of the exemplary embodiments. The present invention is related to oral pharmaceutical compositions comprising FA-aa's suitable for increasing the bioavailability of therapeutic macromolecules (e.g. peptides and proteins) and their absorption.
One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule, such as hydrophilic peptides or proteins and at least one FA-aa. One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is ahydrophilic peptide or protein.
This invention also relates to a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is a peptide.
This invention also relates to a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is a therapeutic active peptide.
One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is a protein.
One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is a therapeutic protein.
One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is an insulin.
One embodiment of the invention is a pharmaceutical composition comprising at least one therapeutic macromolecule and at least one FA-aa, wherein said therapeutic macromolecule is an insulin peptide.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid, said nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms, said one or more nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a polar uncharged amino acid.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a polar uncharged amino acid, said polar uncharged amino acid may be selected from the group consisting of Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin).
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a polar acidic amino acid.
In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and one or more FA-aa, based on a polar acidic amino acid, said polar acidic amino acid may be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu). In one embodiment of the invention, the pharmaceutical composition comprises at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and FA-aa's, based on a mixture FA-aa's.
In one embodiment a pharmaceutical composition according to the present invention comprises one or more commercially available FA-aa's.
According to this invention a FA-aa comprises an amino residue and a fatty acid attached to the amino acid by acylation of said amino acid's alpha-amino group.
In one embodiment, an amino acid residue according to this invention includes the form of its free acid or a salt.
In one embodiment an amino acid residue according to this invention includes the form of its free acid or sodium (Na+) salt.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid and a fatty acid moiety consisting of 10 carbon atoms.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid and a fatty acid moiety consisting of 12 carbon atoms.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid and a fatty acid moiety consisting of 14 carbon atoms.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid and a fatty acid moiety consisting of 16 carbon atoms.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid and a fatty acid moiety consisting of 18 carbon atoms.
In one embodiment a FA-aa comprises an amino acid residue acylated with a fatty acid or salt thereof.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is located at the alpha amino group of the amino acid.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 8 to 18 carbon atoms.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 10 carbon atoms. In one
embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 12 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 14 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 16 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is consisting of 18 carbon atoms.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is in the form of its free acid or salt. In one embodiment a FA-aa according to this invention comprises an acylated amino acid, wherein the fatty acid moiety is in the form of its free acid or sodium (Na+) salt. In one embodiment a FA-aa according to this invention comprise amino acid residues in the form of their free acid or a salt. In one embodiment a FA-aa according to this invention comprises amino acid residues in the form of their free acid or a sodium (Na+) salt. In one embodiment a FA-aa according to this invention is soluble at intestinal pH values, particularly in the 5.5 to 8.0 range. In one embodiment a FA-aa according to this invention is soluble at intestinal pH values, particularly in the 6.5 to 7.0 range.
In one embodiment a FA-aa according to this invention has a solubility of at least
5mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 10mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 20mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 30mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 40 mg/mL. In one embodiment a FA-aa according to this invention has a solubility of at least 50mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 60mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 70mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 80mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 90mg/ml_. In one embodiment a FA-aa according to this invention has a solubility of at least 100mg/ml_.
In one embodiment a FA-aa according to this invention has a solubility of at least 5mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 10mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 20mg/ml_in water. In one embodiment a FA-aa according to this invention has a solubility of at least 30mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 40 mg/mL in water. In one embodiment a FA-aa according to this invention has a solubility of at least 50mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 60mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 70mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 80mg/ml_in water. In one embodiment a FA-aa according to this invention has a solubility of at least 90mg/ml_ in water. In one embodiment a FA-aa according to this invention has a solubility of at least 100mg/ml_ in water.
A FA-aa according to the present invention may be represented by the general formula A-X, wherein A is an amino acid residue, based on a non-cationic amino acid and X is a fatty acid attached by acylation to A's alpha amino group.
In one embodiment a FA-aa according to the present invention may be represented by the general formula A-X, wherein A is an amino acid residue, based on a nonpolar hydrophobic amino acid and X is a fatty acid attached by acylation to A's alpha amino group.
In one embodiment a FA-aa according to the present invention may be represented by the general formula A-X, wherein A is an amino acid residue, based on a polar uncharges amino acid and X is a fatty acid attached by acylation to As alpha amino group.
In one embodiment a FA-aa according to the present invention may be represented by the general formula A-X, wherein A is an amino acid residue, based on a polar acidic amino acid and X is a fatty acid attached by acylation to As alpha amino group.
A FA-aa according to the present invention may be represented by the general formula;
Figure imgf000013_0001
wherein R1 is a fatty acid chain comprising between from 8 to 18 carbons, R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), R3 is either H, or a salt thereof, and R4 is an amino acid side chain of a non-cationic amino acid.
In one embodiment a FA-aa according to the present invention may be represented by the general formula:
Figure imgf000013_0002
wherein R1 is a fatty acid chain comprising 8 to 18 carbon atoms, R2 is either H (i.e.
hydrogen) or CH3 (i.e. methyl group), R3 is either H, or a sodium salt (Na+) thereof, and R4 is a amino acid side chain of a non-cationic amino acid.
In one embodiment a FA-aa according to this invention may be chosen from the group consisting of formula (a), (b), (c), (d), (e), (f), (g), (h), (i), (j), (k), (I), (m), (n), (o), (p), (q) or (r), wherein R1 is a fatty acid chain comprising between from 8 to 18 carbons, R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), and R3 is either H, or a salt thereof.
In one embodiment a FA-aa according to this invention may be chosen from the group consisting of formula (a), (b), (c), (d), (e), (f), (g), (h), (i), (j), (k), (I), (m), (n), (o), (p), (q) or (r), wherein R1 is a fatty acid chain comprising 8 to 18 carbon atoms, R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), and R3 is either H, or a sodium (Na+) salt thereof.
Figure imgf000014_0001
Figure imgf000014_0002
(c),
Figure imgf000015_0001
(d),
Figure imgf000015_0002
Figure imgf000015_0003
Figure imgf000015_0004
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000017_0002
Figure imgf000017_0003
Figure imgf000017_0004
An amino acid residue according to this invention may be based on a non-cationic amino acid.
An amino acid residue according to this invention may be based on a non-cationic amino acid, said non-cationic amino acid may be selected from the group consisting nonpolar hydrophobic amino acids, polar uncharged amio acids and polar acidic amino acids.
An amino acid residue according to this invention may be based on a non-cationic amino acid, said non-cationic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
In one embodiment an amino acid residue of a FA-aa according to this invention may be based on a nonpolar hydrophobic amino acid.
In one embodiment an amino acid residue of a FA-aa according to this invention may be based on a nonpolar hydrophobic amino acid, said nonpolar hydrophobic amino acid may be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment a FA-aa can be selected from the group consisting of: Sodium capric alaninate, N-decanoyl-L-alanine, Sodium capric isoleucinate, N-decanoyl-L-isoleucine, Sodium capric leucinate, N-decanoyl-L-leucine, Sodium capric methioninate, N-decanoyl-L- methionine, Sodium capric phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric prolinate, N-decanoyl-L-proline, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L-tryptophane, Sodium capric valinate, N-decanoyl-L- valine, Sodium capric sarcosinate and N-decanoyl-L-sarcosine.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro) and Sarcosinate.
In one embodiment a FA-aa can be selected from the group consisting of: Sodium lauroyl alaninate, N-dodecanoyl-L-alanine, Sodium lauroyl isoleucinate, N-dodecanoyl-L- isoleucine, Sodium lauroyl leucinate, N-dodecanoyl-L-leucine, , Sodium lauroyl methioninate, N-dodecanoyl-L-methionine, Sodium lauroyl phenylalaninate, N-dodecanoyl-L-phenylalanine, Sodium lauroyl prolinate, N-dodecanoyl-L-proline, Sodium lauroyl tryptophanate, N- dodecanoyl-L-tryptophane, Sodium lauroyl valinate, N-dodecanoyl-L-valine, Sodium lauroyl sarcosinate, N-dodecanoyl-L-sarcosine, Sodium lauroyl sarcosinate, Sodium oleoyl sarcosinate and Sodium N-decyl leucine.
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 8 to 18 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment a FA-aa can be selected from the group consisting of: Sodium capric alaninate, N-decanoyl-L-alanine, Sodium capric leucinate, N-decanoyl-L-leucine, Sodium capric phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric valinate, N- decanoyl-L-valine, Sodium N-decyl leucine,
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms. In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a nonpolar hydrophobic amino acid and a fatty acid moiety consisting of 18 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Phenylalanine (Phe).
In one embodiment a FA-aa can be selected from the group consisting of: Sodium lauroyl alaninate, N-dodecanoyl-L-alanine, Sodium lauroyl leucinate, N-dodecanoyl-L- leucine, Sodium lauroyl phenylalaninate, N-dodecanoyl-L-phenylalanine, Sodium lauroyl valinate, N-dodecanoyl-L-valine, In one embodiment an amino acid residue of a FA-aa according to this invention can be based on a polar uncharged amino acid.
In one embodiment an amino acid residue of a FA-aa according to this invention can be based on a polar uncharged amino acid, said polar uncharged amino acid can be selected from the group consisting of Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin).
In one embodiment a FA-aa can be selected from the group consisting of: Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L- aspartic acid, Sodium lauroyl cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyl glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyl glycinate, N-dodecanoyl-L-glycine, Sodium lauroyl serinate, N-dodecanoyl-L-serine, Sodium lauroyl threoninate, N-dodecanoyl- L-threonine, Sodium lauroyl tyrosinate, N-dodecanoyl-L-tyrosine, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N-decanoyl-L-cysteine, Sodium capric glutaminate, N- decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tyrosinate and N-decanoyl-L-tyrosine, Sodium lauroyl asparaginate, N-dodecanoyl-L- asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyl glutaminate, N-dodecanoyl-L- glutamine, Sodium lauroyl glycinate, N-dodecanoyl-L-glycine, Sodium lauroyl serinate, N- dodecanoyl-L-serine, Sodium lauroyl threoninate, N-dodecanoyl-L-threonine, Sodium lauroyl tyrosinate, N-dodecanoyl-L-tyrosine, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N- decanoyl-L-cysteine, Sodium capric glutaminate, N-decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tyrosinate and N-decanoyl-L-tyrosine.
In one embodiment an amino acid residue of a FA-aa according to this invention can be based on a polar acidic amino acid.
In one embodiment an amino acid residue of a FA-aa according to this invention can be based on a polar acidic amino acid, said polar acidic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 10 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 12 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu). In one embodiment a FA-aa can be selected from the group consisting of: Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N- dodecanoyl-L-aspartic acid, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N- decanoyl-L-aspartic acid, Sodium capric glutamic acid and N-decanoyl-L-glutamic acid.
In one embodiment a FA-aa can be selected from the group consisting of: Amisoft HS-1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium CocoyI Glutamate) and Sodium cocoyl glutamate, Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric glutamic acid and N-decanoyl-L-glutamic acid.
In one embodiment a FA-aa can be selected from the group consisting of: Amisoft HS-1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)),
Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium Cocoyl Glutamate) and Sodium cocoyl glutamate.
In one embodiment an amino acid residue of a FA-aa according to this invention may be based on a polar acidic amino acid.
In one embodiment an amino acid residue of a FA-aa according to this invention may be based on a polar acidic amino acid, said polar acidic amino acid may be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 14 carbon atoms, said nonpolar hydrophobic amino acid may be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 16 carbon atoms, said nonpolar hydrophobic amino acid may be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu).
In one embodiment a FA-aa according to this invention comprises an acylated amino acid based on a polar acidic amino acid and a fatty acid moiety consisting of 18 carbon atoms, said nonpolar hydrophobic amino acid can be selected from the group consisting of Aspartic acid (Asp) and Glutamic acid (Glu). In one embodiment a FA-aa may be selected from the group consisting of: Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N- dodecanoyl-L-aspartic acid, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N- decanoyl-L-aspartic acid, Sodium capric glutamic acid and N-decanoyl-L-glutamic acid.
In accordance with the present invention an amino acid amino acid may be selected from the group constsiting of Amisoft HS-1 1 P (Sodium StearoyI Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium Cocoyl Glutamate) and Sodium cocoyl glutamate.
In accordance with the present invention an amino acid amino acid FA-aa may be selected from the group consisting of: Sodium lauroyl asparaginate, N-dodecanoyl-L- asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium capric asparaginate, N-decanoyl-L- asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric glutamic acid and N-decanoyl-L-glutamic acid.
In accordance with the present invention an amino acid amino acid may be selected from the group constsiting of Amisoft HS-1 1 P (Sodium StearoyI Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium Cocoyl Glutamate) and Sodium cocoyl glutamate.
In one embodiment an amino acid moiety of a FA-aa according to this invention is an amino acids that are not encoded by the genetic code.
In one embodiment an amino acid moiety of a FA-aa according to this invention is Sarcosinate.
In one embodiment an amino acid residue of a FA-aa according to this invention is an free acid or salt form of an amino acid that are not encoded by the genetic code.
In one embodiment an amino acid residue of a FA-aa according to this invention is the free acid or salt form of Sarcosinate.
In one embodiment an amino acid moiety of a FA-aa according to this invention is selected from the group comprising Leucine and Phenylalanine.
Modifications of amino acids by acylation are readily performed using acylation agents known in the art that react with the free alpha-amino group of the amino acid.
The following FA-aa's are commercially available:
Brand name Chemical name Provider (2011 -04-14)
Hamposyl L-95 Sodium lauroyl sarcosine Chattem Chemicals
Hamposyl O Sodium oleoyl sarcosine Chattem Chemicals
Hamposyl C Sodium cocoyl sarcosine Chattem Chemicals Brand name Chemical name Provider (2011 -04-14)
Hamposyl L-30 Sodium lauroyl sarcosine Chattem Chemicals
Amisoft HS-1 1 P Sodium stearoyl glutamate Ajinomoto
Amisoft LS-1 1 Sodium lauroyl glutamate Ajinomoto
Amisoft CS-1 1 Sodium cocoyl glutamate Ajinomoto
Amisoft MS-1 1 Sodium myristoyl glutamate Ajinomoto
Amilite GCS-1 1 Sodium cocoyl glycinate Ajinomoto
According to the present invention, the FA-aa may be part of an oral pharmaceutical composition.
In one embodiment of the invention the pharmaceutical composition comprises of at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and at least on FA-aa and propylene glycol.
In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system, such as SEDDS,
SMEDDS or SNEDDS. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system, such as SEDDS, SMEDDS or SNEDDS. Liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention may be encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form. Thus the term "solid" as used herein refers to liquid compositions encapsulated in a soft or hard capsule technology, but also to tablets and multiparticulates.
Liquid or semisolid SEDDS, SMEDDS or SNEDDS according to the invention may be encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form which may further comprise enteric or delayed release coatings.
Liquid or semisolid SEDDS, SMEDDS or SNEDDS comprising FA-aa's according to the invention may be encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form which may further comprise enteric or delayed release coatings, such as poly(meth)acrylates, commercially known as Eudragit®.
In one embodiment of the invention the pharmaceutical composition is a SEDDS,
SMEDDS or SNEDDS, comprising at least one therapeutic macromolecule, such as a hydrophilic peptide or protein and at least one FA-aa, propylene glycol.
In one embodiment the pharmaceutical composition according to the present comprises less than 10% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 9% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 8% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 7% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 6% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 5% (w/w) water. In one
embodiment the pharmaceutical composition according to the present comprises less than 4% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 3% (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 2% (w/w) water. In one
embodiment the pharmaceutical composition according to the present comprises less than 1 % (w/w) water. In one embodiment the pharmaceutical composition according to the present comprises less than 0% (w/w) water.
In one embodiment the pharmaceutical composition according to the present invention is is a liquid. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 10% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 9% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 8% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 7% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 6% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 5% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 4% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 3% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 2% (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 1 % (w/w) water. In one embodiment the pharmaceutical composition according to the present invention is is a liquid and comprises less than 0% (w/w) water.
In one embodiment of the invention the pharmaceutical composition comprises at least one therapeutic macromoecule. In one embodiment a therapeutic macromolecule, such as a hydrophilic peptide or protein according to this invention is a therapeutic active peptide or protein. In one embodiment a therapeutic peptide or protein according to this invention is a hydrophilic peptide or protein.
In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 50mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least
60mg/ml_ in water In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 70mg/ml_ in water In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 80mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 90mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 100mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 1 10mg/mL in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 120mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 130mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 140mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 150/mL in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 160mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 170mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 180mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 190mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 200mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 210mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 220mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 230mg/ml_ in water. In one embodiment a hydrophilic peptide or protein of this invention is a peptide or protein having a solubility of at least 240mg/ml_ in water.
In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 1500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 1750Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 2000Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 2250Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 2500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 2750Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 3000Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 3250Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 3500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 3750Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 4000Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 4250Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 4500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 4750Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 5000Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of more than 1500Da. In one embodiment a therapeutic active peptide or protein according to this invention is a peptide or protein of between 1500Da and 5000Da.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent .
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the solvent is selected from the group consisting of water and propylene glycol.
in one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, wherein said Polyethylene glycol sorbitan fatty acid ester is selected from the group consisting of Tween 20, Tween 21 , Tween 40, Tween 60, Tween 65, Tween 80, Tween 81 and Tween 85. In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, wherein said Polyethylene glycol sorbitan fatty acid ester is selected from the group consisting of Tween 20, Tween 21 , Tween 40, Tween 60, Tween 65, Tween 80, Tween 81 and Tween 85.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent, wherein the solvent is selected from the group consistin g of water and propylene glycol.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the Polyethylene glycol sorbitan fatty acid ester is a Polyethylene glycol sorbitan trioleate, commercially known as Tween 85.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the Polyethylene glycol sorbitan fatty acid ester is a Polyethylene glycol sorbitan trioleate, commercially known as Tween 85 and the solvent is selected form the group consisting of water and propylene glycol.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan trioleate commercially known as Tween 85 and a polar or semipolar solvent selected from the group consistin of water and propylene glycol, wherein the composition forms a microemulsion after dilution in an aqeous medium. In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan trioleate commercially known as Tween 85 and a polar or semipolar solvent selected from the group consistin of water and propylene glycol, wherein the composition forms a microemulsion after dilution in an aqeous medium.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the Polyethylene glycol sorbitan fatty acid ester is a Polyethylene glycol sorbitan trioleate, commercially known as Tween 20.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester, and a polar or semipolar solvent , wherein the Polyethylene glycol sorbitan fatty acid ester is a Polyethylene glycol sorbitan monolaurate, commercially known as Tween 20 and the solvent is selected form the group consisting of water and propylene glycol.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan monolaurate commercially known as Tween 20 and a polar or semipolar solvent selected from the group consistin of water and propylene glycol, wherein the composition forms a microemulsion after dilution in an aqeous medium.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan monolaurate commercially known as Tween 20 and a polar or semipolar solvent selected from the group consistin of water and propylene glycol, wherein the composition forms a microemulsion after dilution in an aqeous medium.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent. In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising
Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent, wherein said polar or semipolar solvent is selected from the group consisting of water and propylene glycol. In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising Polyethylene glycol sorbitan fatty acid ester and a polar or semipolar solvent, wherein said polar or semipolar solvent is selected from the group consisting of water and propylene glycol.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester and a polar or semipolar solvent (such as water or propylene glycol). In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester (Span 10, 20, 40, 60 or 80), and a polar or semipolar solvent (such as water or propylene glycol).
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester, wherein said sorbitan fatty acid ester is selected form the group consisting of Span 10, Span 20, Span 40, Span 60 and Span 80. In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester, wherein said sorbitan fatty acid ester is selected form the group consisting of sorbitan laurate commercially known as Span 20, sorbitan mono palmitate commercially known as Span 40, sorbitan mono stearate commercially known as Span 60 and sorbitan oleate commercially known as Span 80.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester and a polar or semipolar solvent. In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester and a polar or semipolar solvent.
In one embodiment a pharmaceutical composition according to the present invention is a liquid and comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one polyglycerol fatty acid ester, further comprising sorbitan fatty acid ester and a polar or semipolar solvent, wherein said polar or semipolar solvent is selected from the group consisting of water or propylene glycol.
In one embodiment of the invention the pharmaceutical composition comprises at least one therapeutic active peptide or protein.
In one embodiment of the present invention the pharmaceutical composition comprises at least one therapeutic active peptide or protein, which has been pH neutralised.
In one embodiment of the invention the therapeutical active peptide or protein is dissolved and the pH of the resulting solution is adjusted to a value of the target pH value, which is 1 unit, alternatively 2 units and alternatively 2.5 pH units above or below the pi of the insulin peptide, whereafter said resulting solution is freeze or spray dryed. In one
embodiment said pH adjustment is performed with a non-volitale acid or base.
In one embodiment of the invention the pharmaceutical composition comprises of at least one insulin peptide and at least on FA-aa. In one embodiment of the invention the pharmaceutical composition comprises of at least one peptide or protein and at least on FA- aa.
In one embodiment of the invention the pharmaceutical composition comprises of at least one insulin peptide and at least on FA-aa and propylene glycol.
In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 10% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 9% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 8% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 7% (w/w) water. In one embodiment the amino acid FA- aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 6% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 5% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 4% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 3% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 2% (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 1 % (w/w) water. In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system comprising less than 0% (w/w) water.
In one embodiment a pharmaceutical composition according to the present invention comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one high HLB surfactant, at least one low HLB co-surfactant and a polar solvent. In one embodiment a pharmaceutical composition according to the present invention comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least one high HLB surfactant, at least one low HLB co- surfactant and a polar solvent.
In one embodiment a pharmaceutical composition according to the present invention comprises at least one therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least two high HLB surfactants, and a polar solvent. In one embodiment a pharmaceutical composition according to the present invention comprises a therapeutic hydrophilic protein or polypeptide, at least one fatty acid acylated amino acid, at least two high HLB surfactants, and a polar solvent.
In one embodiment the amino acid FA-aa may be used in a liquid or semisolid liquid and surfactant based delivery system, such as SEDDS, SMEDDS or SNEDDS. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 10% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 9% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 8% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 7% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 6% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 6% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 5% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 4% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 3% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 2% (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 1 % (w/w) water. In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system comprising less than 0% (w/w) water.
In one embodiment the amino acid FA-aa may be used in a solid surfactant based delivery system, such as SEDDS, SMEDDS or SNEDDS.
In one embodiment the pharmaceutical composition according to the present invention is a liquid.
In one embodiment pharmaceutical composition is a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form. In one embodiment a soft capsule technology used for encapsulating a composition according to the present invention is gelatine free. In one embodiment a gelatine free soft capsule technology as commercially known under the name Vegicaps® from Catalent® is used for encapsulation of the pharmaceutical composition according to the present invention.
In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 10% (w/w) water. In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 9% (w/w) water In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 8% (w/w) water. In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS
comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 7% (w/w) water In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 6% (w/w) water. In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 5% (w/w) water. In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 4% (w/w) water. In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 3% (w/w) water. In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 2% (w/w) water. In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 1 % (w/w) water In one embodiment the pharmaceutical composition a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention and is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form comprising less than 0% (w/w) water.
In one embodiment a liquid or semisolid formulation according to the invention is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form further comprising an enteric or delayed release coatings. In one embodiment a liquid or semisolid formulation according to the invention is encapsulated with any available enteric soft- or hard capsule technology to result in a solid oral pharmaceutical dosage.
In one embodiment a liquid or semisolid SEDDS, SMEDDS or SNEDDS
comprisning FA-aa's according to the invention is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form further comprising an enteric or delayed release coatings. In one embodiment a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprisning FA-aa's according to the invention is
encapsulated with any available enteric soft- or hard capsule technology to result in a solid oral pharmaceutical dosage.
In one embodiment a liquid or semisolid SEDDS, SMEDDS or SNEDDS comprising FA-aa's according to the invention is encapsulated with any available soft- or hard capsule technology to result in a solid oral pharmaceutical dosage form which may further comprise an enteric or delayed release coatings, such as poly(meth)acrylates, commercially known as Eudragit®.
In one embodiment, the coating comprises at least one release modifying polymer which can be used to control the site where the drug (insulin derivative) is released. The modified release polymer can be a polymethacrylate polymer such as those sold under the Eudragit® trade name (Evonik Rohm GmbH, Darmstadt, Germany), for example Eudragit® L30 D55, Eudragit® L100-55, Eudragit® L100, Eudragit® S100, Eudragit® S12.5,
Eudragit® FS30D, Eudragit® NE30D and mixtures thereof as e.g. described in Eudragit® Application Guidelines, Evonik Industries, 1 1 th edition, 09/2009.
In one embodiment of the invention the pharmaceutical composition is a formulation, comprising at least one insulin and at least one FA-aa, propylene glycol.
In one embodiment of the invention the pharmaceutical composition comprises of at least one insulin and at least one FA-aa, propylene glycol.
In one embodiment of the invention the pharmaceutical comprises at least one peptide or protein and at least one FA-aa, propylene glycol.
In one embodiment of the invention the pharmaceutical composition is a SEDDS, SMEDDS or SNEDDS, comprising at least one peptide or protein and at least one FA-aa, propylene glycol.
The components of the drug delivery system may be present in any relative amounts. In one embodiment the drug delivery system comprises up to 90% of a surfactant, or up to 90% of a polar organic solvent such as Polyethylene glycol (PEG) 300 g/mol, PEG 400 g/mol, PEG 600 g/mol, PEG 1000 g/mol, or up to 90% of a lipid component. PEGs are prepared by polymerization of ethylene oxide and are commercially available over a wide range of molecular weights from 300 g/mol to 10,000,000 g/mol.
In one embodiment the oral pharmaceutical composition comprises from 5 to 20% of propylene glycol.
In one embodiment, the oral pharmaceutical composition comprises at least one FA- aa, propylene glycol, and at least two non ionic surfactants.
In one embodiment, the oral pharmaceutical composition comprises at least one FA- aa, propylene glycol, polysorbate 20 and a co-surfactant. Polysorbate 20 is a polysorbate surfactant whose stability and relative non-toxicity allows it to be used as a detergent and emulsifier in a number of domestic, scientific, and pharmacological applications. The number 20 refers to the total number of oxyethylene -(CH2CH20)- groups found in the molecule.
In one embodiment of the present invention, the oral pharmaceutical composition comprises at least one FA-aa, propylene glycol, polysorbate 20 and a polyglycerol fatty acid ester.
In one embodiment, the oral pharmaceutical composition comprises at least one FA- aa, propylene glycol, polysorbate 20 and a co-surfactant.
In one embodiment, the oral pharmaceutical composition comprises at least one FA- aa, propylene glycol, polysorbate 20 and a polyglycerol fatty acid ester such as diglycerol monocaprylate.
In certain embodiments of the present invention, the pharmaceutical composition may comprise additional excipients commonly found in pharmaceutical compositions, examples of such excipients include, but are not limited to, antioxidants, antimicrobial agents, enzyme inhibitors, stabilizers, preservatives, flavors, sweeteners and other components as described in Handbook of Pharmaceutical Excipients, Rowe et al., Eds., 4th Edition,
Pharmaceutical Press (2003), which is hereby incorporated by reference
These additional excipients may be in an amount from about 0.05-5% by weight of the total pharmaceutical composition. Antioxidants, anti-microbial agents, enzyme inhibitors, stabilizers or preservatives typically provide up to about 0.05-1 % by weight of the total pharmaceutical composition. Sweetening or flavouring agents typically provide up to about 2.5% or 5% by weight of the total pharmaceutical composition.
Oral pharmaceutical compositions according to this invention may be formulated as solid dosage forms.
Oral pharmaceutical compositions according to this invention may be formulated as solid dosage forms and may be selected from the group consisting of capsules, tablets, dragees, pills, lozenges, powders and granules. Oral pharmaceutical compositions according to this invention may be formulated as mutliparticulate dosage forms.
Oral pharmaceutical compositions according to this invention may be formulated as mutliparticulate dosage forms and may be selected from the group consisting of pellets, microparticles, nanoparticles, liquid or semisolid fill formulations in soft- or hard capsules, enteric coated soft- hard capsules.
In one embodiment the oral pharmaceutical compositions may be prepared with one or more coatings such as enteric coatings or be formulated as delayed release formulations according to methods well known in the art.
Enteric or delayed release coatings according to this invention may be based on
poly(meth)acrylates commercially known as Eudragit®.
In one embodiment, the pharmaceutical composition according to the invention is used for the preparation of a medicament.
In one embodiment, the pharmaceutical composition according to the invention is used for the preparation of a medicament for the treatment or prevention of hyperglycemia, type 2 diabetes mellitus, impaired glucose tolerance, type 1 diabetes mellitus and/or anti obesity treatment.
The terms "fatty acid N-acylated amino acid" or "acylated amino acid" may be used interchangeable and refer when used herein to an amino acids that is acylated with a fatty acid at its alpha-amino group.
Amino acids exist in the stereoisomeric form of either D (dextro) or L (levo). The D and L refer to the absolute confirmation of optically active compounds. With the exception of glycine, all other amino acids are mirror images that can not be superimposed. Most of the amino acids found in nature are of the L-type. Hence, eukaryotic proteins are always composed of L-amino acids although D-amino acids are found in bacterial cell walls and in some peptide antibiotics. At least 300 amino acids have been described in nature but only twenty of these are typically found as components in human peptides and proteins. Twenty standards amino acids are used by cells in peptide biosynthesis, and these are specified by the general genetic code. The twenty standard amino acids are Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Methionine (Met), Proline (Pro), Apartic acid (Asp), Gltamic acid (Glu), Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Apsaragine (Asn), Glutamine (Gin), Lysine (Lys), Arginine (Arg) and Histidine (His).
The amino acid moiety of the modified FA-aa may be in the form of a pure enantiomer wherein the stereo configuration of the chiral amino acid moiety is either D or L (or if using the R/S terminology: either R or S) or it may be in the form of a mixture of enantiomers (D and L / R and S).
In one embodiment of the invention the amino acid moiety is in the form of a mixture of enantiomers.
In one embodiment the amino moiety is in the form of a pure enantiomer. In one embodiment the chiral amino acid moiety is in the L form. In one embodiment the chiral amino acid moiety is in the D form.
As used herein the term "non-cationic amino acid" shall be understood as referring to any amino acid selected from the group consisting of nonpolar hydrophobic amino acids, polar uncharged amino acids and polar acidic amino acids.
The term "nonpolar hydrophobic amino acids" as used herein refer to catogorisation of amino acids used by the person skilled in the art. The term "polar uncharged amino acids" as used herein refer to catogorisation of amino acids used by the person skilled in the art. The term "and polar acidic amio acids" as used herein refer to catogorisation of amino acids used by the person skilled in the art. As used herein the term "non-cationic amino acid" comprises the following amino acids: Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
With the term "oral bioavailability" is herein meant the fraction of the administered dose of drug that reaches the systemic circulation after having been administered orally. By definition, when a medication is administered intravenously, its bioavailability is 100%.
However, when a drug is administered orally the bioavailability of the active ingredient decreases due to incomplete absorption and first-pass metabolism. The biological activity of an insulin peptide may be measured in an assay as known by a person skilled in the art as e.g. described in WO 2005012347.
The term "surfactant" as used herein refers to any substance, in particular a detergent, that can adsorb at surfaces and interfaces, such as but not limited to liquid to air, liquid to liquid, liquid to container or liquid to any solid and which has no charged groups in its hydrophilic groups.
The term "permeation enhancer" when used herein refers to biologicals or chemicals that promote the absorption of drugs.
The term "preservative" as used herein refers to a chemical compound which is added to a pharmaceutical composition to prevent or delay microbial activity (growth and metabolism). Examples of pharmaceutically acceptable preservatives are phenol, m-cresol and a mixture of phenol and m-cresol.
The term "macromolecular" or "macromolecule" used herein refer to non-polymeric molecules and comprises nucleic acids, peptides, proteins, carbohydrates, and lipids.
The term "polypeptide" and "peptide" as used herein means a compound composed of at least two constituent amino acids connected by peptide bonds. The constituent amino acids may be from the group of the amino acids encoded by the genetic code and they may be natural amino acids which are not encoded by the genetic code, as well as synthetic amino acids. Commonly known natural amino acids which are not encoded by the genetic code are e.g., γ-carboxyglutamate, ornithine, phosphoserine, D-alanine and D-glutamine. Commonly known synthetic amino acids comprise amino acids manufactured by chemical synthesis, i.e. D-isomers of the amino acids encoded by the genetic code such as D-alanine and D-leucine, Aib (a-aminoisobutyric acid), Abu (a-aminobutyric acid), Tie (tert-butylglycine), β-alanine, 3-aminomethyl benzoic acid, anthranilic acid.
The term "Protein" as used herein means a biochemical compound consisting of one or more polypeptides.
The term "macromolecular therapeutic" or "therapeutic macromolecule" may be used interchangeable and as used herein refer to nucleic acids, peptides, proteins, carbohydrates, and lipids as well as non-polymeric molecules with large molecular mass used in therapy and includes without being limited therto insulin, insulin analouges and insulin derivatives. In one embodiment lage molecular mass means a molecular mass above 1500Da. In one embodiment large molecular mass means a molecular mass between 150Da and 6000Da.
The term "drug", "therapeutic", "medicament" or "medicine" when used herein refer to an active ingredient used in a pharmaceutical composition, which may be used in therapy and thus also refer to what was defined as "macromolecular therapeutic" or "therapeutic macromolecule" in the present patent application.
With "insulin peptide", "an insulin peptide" or "the insulin peptide" as used herein is meant human insulin comprising disulfide bridges between CysA7 and CysB7 and between CysA20 and CysB19 and an internal disulfide bridge between CysA6 and CysA1 1 or an insulin analogue or derivative thereof.
The term "peptide" as used herein comprises also peptides, proteins, conjugates of such peptides and proteins and biologically active fragments thereof. The term "protein" comprises peptides and also refers to proteins and biologically active fragments thereof. Human insulin consists of two polypeptide chains, the A and B chains which contain 21 and 30 amino acid residues, respectively. The A and B chains are interconnected by two disulphide bridges. Insulin from most other species is similar, but may contain amino acid substitutions in some positions.
The term "insulin" as used herein is, if not specified further, an insulin selected from the group consisting of human insulin, insulin analogues and insulin derivatives.
An insulin analogue as used herein is a polypeptide, such as a insulin peptide which has a molecular structure which formally may be derived from the structure of a naturally occurring insulin, for example that of human insulin, by deleting and/or substituting at least one amino acid residue occurring in the natural insulin and/or by adding at least one amino acid residue.
The term "insulin analogue" as used herein means a modified insulin wherein one or more amino acid residues of the insulin have been substituted by other amino acid residues and/or wherein one or more amino acid residues have been deleted from the insulin and/or wherein one or more amino acid residues have been added and/or inserted to the insulin.
In one embodiment an insulin analogue according to the invention comprises less than 8 modifications (substitutions, deletions, additions) relative to human insulin.
In one embodiment an insulin analogue comprises less than 7 modifications (substitutions, deletions, additions) relative to human insulin. In one embodiment an insulin analogue comprises less than 6 modifications (substitutions, deletions, additions) relative to human insulin.
In one embodiment an insulin analogue comprises less than 5 modifications (substitutions, deletions, additions) relative to human insulin. In one embodiment an insulin analogue comprises less than 4 modifications (substitutions, deletions, additions) relative to human insulin. In one embodiment an insulin analogue comprises less than 3 modifications (substitutions, deletions, additions) relative to human insulin. In one embodiment an insulin analogue comprises less than 2 modifications (substitutions, deletions, additions) relative to human insulin.
The term "insulin derivative" as used herein refers to chemically modified parent insulin or an analogue thereof, wherein the modification(s) are in the form of attachment of amides, carbohydrates, alkyl groups, acyl groups, esters, PEGylations, and the like.
An insulin derivative according to the invention is a naturally occurring insulin or an insulin analogue which has been chemically modified, e.g. by introducing a side chain in one or more positions of the insulin backbone or by oxidizing or reducing groups of the amino acid residues in the insulin or by converting a free carboxylic group to an ester group or to an amide group. Other derivatives are obtained by acylating a free amino group or a hydroxy group, such as in the B29 position of human insulin or desB30 human insulin.
Herein, the term "acylated insulin" covers modification of insulin by attachment of one or more lipophilic substituents optionally via a linker to the insulin peptide.
An insulin derivative is thus human insulin, an insulin analogue or insulin peptide which comprises at least one covalent modification such as a side-chain attached to one or more amino acids of the insulin peptide.
Herein, the naming of the insulin peptide is done according to the following principles: The names are given as mutations and modifications (acylations) relative to human insulin. For the naming of the acyl moiety, the naming is done according to lUPAC nomenclature and in other cases as peptide nomenclature. For example, naming the acyl moiety:
Figure imgf000041_0001
may be e.g. "octadecanedioyl-Y-L-Glu-OEG-OEG", or "17-carboxyheptadecanoyl-Y-L-Glu- OEG-OEG", wherein OEG is short hand notation for the amino acid -
NH(CH2)20(CH2)20CH2CO-, and γ-L-Glu (or g-L-Glu) is short hand notation for the L-form of the amino acid gamma glutamic acid moiety.
The acyl moiety of the modified peptides or proteins may be in the form of a pure enantiomer wherein the stereo configuration of the chiral amino acid moiety is either D or L (or if using the R/S terminology: either R or S) or it may be in the form of a mixture of enantiomers (D and L / R and S). In one embodiment of the invention the acyl moiety is in the form of a mixture of enantiomers. In one embodiment the acyl moiety is in the form of a pure enantiomer. In one embodiment the chiral amino acid moiety of the acyl moiety is in the L form. In one embodiment the chiral amino acid moiety of the acyl moiety is in the D form.
In one embodiment an insulin derivative in an oral pharmaceutical composition according to the invention is an insulin peptide that is acylated in one or more amino acids of the insulin peptide.
In one embodiment an insulin derivative in an oral pharmaceutical composition according to the invention is an insulin peptide that is stabilized towards proteolytic degradation (by specific mutations) and further acylated at the B29-lysine. A non-limiting example of insulin peptides that are stabilized towards proteolytic degradation (by specific mutations) may e.g. be found in WO 2008034881 , which is hereby incorporated by reference.
The acylated insulin peptides suitable for this invention may be mono-substituted having only one acylation group attached to a lysine amino acid residue in the protease stabilized insulin molecule.
A non-limiting list of acylated insulin peptides suitable for the liquid oral
pharmaceutical composition of the invention may e.g. be found in WO 2009/1 15469 such as in the passage beginning on page 24 thereof and continuing the next 6 pages.
In one embodiment of the invention, the acylated insulin peptide is selected from the group consisting of:
B29K(N(e)hexadecanedioyl-Y-L-Glu) A14E B25H desB30 human insulin;
B29K(N(e)octadecanedioyl-Y-L-Glu-OEG-OEG) desB30 human insulin;
B29K(N(e)octadecanedioyl-Y-L-Glu) A14E B25H desB30 human insulin;
B29K(N(e)eicosanedioyl-Y-L-Glu) A14E B25H desB30 human insulin;
B29K(N(e)octadecanedioyl-Y-L-Glu-OEG-OEG) A14E B25H desB30 human insulin;
B29K(N(e)eicosanedioyl-Y-L-Glu-OEG-OEG) A14E B25H desB30 human insulin;
B29K(N(e)eicosanedioyl-Y-L-Glu-OEG-OEG) A14E B16H B25H desB30 human insulin;
B29K(N(e)hexadecanedioyl-Y-L-Glu) A14E B16H B25H desB30 human insulin; B29K(N(e)eicosanedioyl-Y-L-Glu-OEG-OEG) A14E B16H B25H desB30 human insulin; and
B29K(N(e)octadecanedioyl) A14E B25H desB30 human insulin.
In one embodiment of the invention, the insulin derivative is
B29K(N(e)octadecanedioyl-Y-L-Glu-OEG-OEG) A14E B25H desB30 human insulin.
A non-limiting list of acylated insulin peptides suitable for the liquid oral
pharmaceutical composition of the invention may e.g. be found in the PCT application WO201 1068019 such as outlined and exemplified in but not limited to the passage beginning on page 20 line 20 and continuing the next 6 pages, to be published in April 2013.
In one embodiment of the invention, the acylated insulin peptide is selected from the group consisting of N-terminally modified insulin consisting of:
A1 (Af.Af-Dimethyl), A14E, Bl tAf.Af-dimethyl), B25H, B29K(/V¾ctadecanedioyl- gGlu-2xOEG), desB30 human insulin
AI CAT.AT-Diethyl), A14E, B1 (AT.AT-diethyl), B25H, B29K(/\fOctadecanedioyl-gGlu- 2xOEG), desB30 human insulin A1 (W.W-Dimethyl), A14E, B1 (/V^/\f-dimethyl), B16H, B25H,
B29K(/\fhexadecanedioyl-gGlu), desB30 human insulin
AI^Ar-Dimethyl), A14E, BI ^.AT-dimethyl), B25H, desB27,
B29K(/\foctadecanedioyl-gGlu), desB30 human insulin
A1 (A/^Af-Dimethyl), A14E, BI ^AT-dimethyl), B25H, desB27,
B29K(/\foctadecanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (Af.Af-Dimethyl), A14E, BI ^/NT-dimethyl), desB27, B29K(Afoctadecanedioyl- gGlu-2xOEG), desB30 human insulin
AI^Ar-Dimethyl), A14E, BI ^AT-dimethyl), B16H, B25H, Β29Κ(ΛΓ
eicosanedioyl -gGlu-2xOEG), desB30 human insulin
AI G^Af-Dimethyl), A14E, B1 F^AF-dimethyl), B25H, desB27,
B29K(/\fhexadecanedioyl-gGlu), desB30 human insulin
AI G^Af-Dimethyl), A14E, B1 F(N(alpha),N(/Va,/Va-dimethyl), B25H, desB27, B29K(/\fhexadecanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (Af./NT-Dimethyl), A14E, B1 (Af.AT-dimethyl), desB27, B29K(Af octadecanedioyl- gGlu), desB30 human insulin
AI ^AT-Dimethyl), A14E, BI ^.AT-dimethyl), B25H, B29K(Afoctadecanedioyl- gGlu), desB30 human insulin
A1 (AfCarbamoyl), A14E, Bl i/NTCarbamoyl), B25H, B29K(/\foctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (/^Carbamoyl), A14E, B1 (/^Carbamoyl), B25H, B29K(/\fhexadecanedioyl-gGlu), desB30 human insulin
A1 (ATCarbamoyl), A14E, Bl ^Carbamoyl), B25H, B29K(Afeicosanedioyl-gGlu), desB30 human insulin
A1 (AfCarbamoyl), A14E, B1 (AfCarbamoyl), B25H, B29K(Afeicosanedioyl-gGlu-
2xOEG), desB30 human insulin
A1 (/^Carbamoyl), A14E, B1 (/^Carbamoyl), B16H, B25H, B29K(/\feicosanedioyl- gGlu-2xOEG), desB30 human insulin
A1 (NaCarbamoyl), A14E, B1 (NaCarbamoyl), B25H, desB27,
B29K(NEoctadecandioyl-gGlu), desB30 human insulin
A1 (NaCarbamoyl), A14E, B1 (NaCarbamoyl), B25H, desB27,
B29K(NEoctadecandioyl-gGlu-2xOEG), desB30 human insulin A1 G(N(alp a)carbamoyl), A14E, B1 F(N(alpha)carbamoyl), desB27, B29K(N(eps)hexadecanedioyl-gGlu), desB30 human insulin
A1 G(N(alpha)carbamoyl), A14E, B1 F(N(alpha)carbamoyl), desB27, B29K(Neps)- hexadecanedioyl-gGlu-2xOEG), desB30 human insulin
A1 G(N(alpha)carbamoyl), A14E, B1 F(N(alpha)carbamoyl), desB27, B29K(Neps)- eicosanedioyl-gGlu), desB30 human insulin
AI GCAfcarbamoyl), A14E, B1 FtAfcarbamoyl), B16H, desB27, B29K(Neps)- eicosanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (/^Carbamoyl), A14E, B1 (/^Carbamoyl), desB27, B29K(A/¾ctadecanedioyl- gGlu), desB30 human insulin
A1 (AfCarbamoyl), A14E, B1 (^Carbamoyl), B16H, B25H, B29K(Afeicosanedioyl- gGlu), desB30 human insulin
A1 (AfCarbamoyl), A14E, Bl tAfCarbamoyl), desB27, B29K(Afoctadecanedioyl- gGlu-2xOEG), desB30 human insulin
A1 (/^Carbamoyl), A14E, Bl tAfcarbamoyl), B25H, B29K(/\foctadecanedioyl-gGlu), desB30 human insulin
A1 (AfCarbamoyl), A14E, B1 (^Carbamoyl), B16H, B25H, B29K(Afeicosanedioyl- gGlu), desB30 human insulin
AI Gi/Nfcarbamoyl), A14E, B1 F^carbamoyl), B25H, desB27,
B29K(/\feicosanedioyl-gGlu-2xOEG), desB30 human insulin
A1 G(Af¾arbamoyl), A14E, B1 FtAfcarbamoyl), desB27, B29K(/\feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
AI G^carbamoyl), A14E, B1 F^carbamoyl), B16H, desB27, Β29Κ(ΛΓ- eicosanedioyl-gGlu-2xOEG), desB30 human insulin
A1 G(/\nhiocarbamoyl), A14E, B1 F(N Afthiocarbamoyl), B25H, desB27, B29K(/\f- octadecanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (AfAcetyl), A14E, BI ^Acetyl), B25H, B29K(/\fhexadecanedioyl-gGlu), desB30 human insulin
A1 (AfAcetyl), A14E, BI ^Acetyl), B25H, desB27, B29K(Afoctadecanedioyl-gGlu), desB30 human insulin
A1 (ATAcetyl), A14E, BI ^Acetyl), B25H, B29K(/\foctadecandioyl-gGlu-2xOEG), desB30 human insulin
A1 (/VDimethylglycyl), A14E, BI ^Dimethylglycyl), B25H, B29K(/\foctadecanedioyl- gGlu-2xOEG), desB30 human insulin A1 (/Va3-(/V,/V-Dimethylamino)propionyl), A14E, Β^Ι ^^Ν,Ν- dimethylamino)propionyl), B25H, B29K(/\foctadecanedioyl-gGlu-2xOEG), desB30 human insulin
AI ^-^/V-Dimethylamino^utanoyl), A14E, Β^Ι ^Λ^Ν,Ν- dimethylamino)butanoyl), B25H, B29K(/\foctadecanedioyl-gGlu-2xOEG), desB30 human insulin
Al i/NTS-il -Piperidiny propionyl), A14E, Bl tAfS-O-piperidinyOpropionyl), B25H, B29K(/\foctadecanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (AfDimethylglycyl), A14E, BI ^Dimethylglycyl), B25H, desB27,
B29K(/\foctadecanedioyl-gGlu), desB30 human insulin
AI GCAfacetyl), A14E, B1 F(/Vaacetyl),B25H, desB27, B29K(/V¾ctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
AI G^-Picolyl), A14E, B1 F(A/tx2-Picolyl), B25H, desB27, B29K(N(eps)- octadecanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (AfAcetyl), A14E, BI ^Acetyl), B25H, B29K(/\feicosanedioyl-gGlu), desB30 human insulin
A1 (AfAcetyl), A14E, BI ^Acetyl), B25H, B29K(/\feicosanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (AfAcetyl), A14E, BI ^Acetyl), B16H, B25H, B29K(/\feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfAcetyl), A14E, BI ^Acetyl), B16H, B25H, B29K(/\feicosanedioyl-gGlu), desB30 human insulin
A1 (AfDimethylglycyl), A14E, BI ^Dimethylglycyl), B16H, B25H,
B29K(/\fhexadecanedioyl-gGlu), desB30 human insulin
A-1 (/VTrimethyl), A14E, B-1 (/VTrimethyl), B25H, B29K(/\foctadecanedioyl-gGlu-
2xOEG), desB30 human insulin
A1 (AfAcetyl), A14E, BI ^Acetyl), desB27, B29K(/\foctadecanedioyl-gGlu), desB30 human insulin
A1 (AfAcetyl), A14E, BI ^Acetyl), desB27, B29K(/\foctadecanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (AfAcetyl), A14E, B'l ihTAcetyl), B25H, B29K(/\foctadecanedioyl-gGlu), desB30 human insulin
AI GCAfAcetyl), A14E, B1 F^Acetyl), desB27, B29K(/\feicosanedioyl-gGlu), desB30 human insulin AI GiAFAcetyl), A14E, B1 F^Acetyl), desB27, B29K(/\feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
AI G^Acetyl), A14E, B1 F^Acetyl), B25H, desB27, B29K(/\feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfSuccinyl), A14E, B1 (Afsuccinyl), B25H, desB27, B29K(/\foctadecanedioyl- gGlu-2xOEG), desB30 human insulin
A1 (AfSuccinyl), A14E, Bl ^succinyl), B25H, B29K(/\foctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfSuccinyl), A14E, Bl ^succinyl), desB27, B29K(/V¾ctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfGlutaryl), A14E, B1 (ATglutaryl), B25H, B29K(/\foctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfGlutaryl), A14E, B1 (ATglutaryl), desB27, B29K(/\foctadecanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfDiglycolyl), A14E, B1 (ΛΡ diglycolyl), B25H, desB27, Β29Κ(ΛΓ octadecanedioyl- gGlu-2xOEG), desB30 human insulin
A1 (AfGlutaryl), A14E, B1 (ATglutaryl), B25H, desB27, B29K(/V¾ctadecanedioyl- gGlu-2xOEG), desB30 human insulin
A1 (AfSuccinyl), A14E, Bl ^succinyl), desB27, B29K(/V¾ctadecanedioyl-gGlu), desB30 human insulin
A1 (AfSuccinyl), A14E, Bl ^succinyl), B25H, desB27, B29K(/\feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfSuccinyl), A14E, Bl ^succinyl), desB27, B29K(/\feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfSuccinyl), A14E, Bl ^succinyl), B16H, desB27, B29K(/\feicosanedioyl-gGlu-
2xOEG), desB30 human insulin
A1 (AfSuccinyl), A14E, Bl ^succinyl), B25H, B29K(/\feicosanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (AfSuccinyl), A14E, Bl ^succinyl), desB27, B29K(/\feicosanedioyl-gGlu), desB30 human insulin
A1 (AfGlutaryl), A14E, B1 (ATglutaryl), desB27, B29K(/\feicosanedioyl-gGlu), desB30 human insulin
A1 (AfGlutaryl), A14E, B1 (ATglutaryl), desB27, B29K(/\feicosanedioyl-gGlu-2xOEG), desB30 human insulin A1 (AfGlutaryl), A14E, B1 (ATglutaryl), B25H, desB27, B29K(/\feicosanedioyl-gGlu- 2xOEG), desB30 human insulin
A1 (AfGlutaryl), A14E, B /Nfglutaryl), desB27, B29K(/\feicosanedioyl-gGlu-2xOEG), desB30 human insulin
A1 (AfGlutaryl), A14E, B1 (Afglutaryl), B25H, B29K(/\feicosanedioyl-gGlu-2xOEG), desB30 human insulin
In one embodiment, an N-terminally modified insulin according to the invention has a peptide part which is selected from the group consisting of the following insulin peptides (i.e. insulins of the invention without N-terminal modifications and without the "lipophilic substituent" or acyl moiety): A14E, B25H, desB30 human insulin; A14H, B25H, desB30 human insulin; A14E, B1 E, B25H, desB30 human insulin; A14E, B16E, B25H, desB30 human insulin; A14E, B25H, B28D, desB30 human insulin; A14E, B25H, B27E, desB30 human insulin; A14E, B1 E, B25H, B27E, desB30 human insulin; A14E, B1 E, B16E, B25H, B27E, desB30 human insulin; A8H, A14E, B25H, desB30 human insulin; A8H, A14E, B25H, B27E, desB30 human insulin; A8H, A14E, B1 E, B25H, desB30 human insulin; A8H, A14E, B1 E, B25H, B27E, desB30 human insulin; A8H, A14E, B1 E, B16E, B25H, B27E, desB30 human insulin; A8H, A14E, B16E, B25H, desB30 human insulin; A14E, B25H, B26D, desB30 human insulin; A14E, B1 E, B27E, desB30 human insulin; A14E, B27E, desB30 human insulin; A14E, B28D, desB30 human insulin; A14E, B28E, desB30 human insulin; A14E, B1 E, B28E, desB30 human insulin; A14E, B1 E, B27E, B28E, desB30 human insulin; A14E, B1 E, B25H, B28E, desB30 human insulin; A14E, B1 E, B25H, B27E, B28E, desB30 human insulin; A14D, B25H, desB30 human insulin; B25N, B27E, desB30 human insulin; A8H, B25N, B27E, desB30 human insulin; A14E, B27E, B28E, desB30 human insulin; A14E, B25H, B28E, desB30 human insulin; B25H, B27E, desB30 human insulin; B1 E, B25H, B27E, desb30 human insulin; A8H, B1 E, B25H, B27E, desB30 human insulin; A8H, B25H, B27E, desB30 human insulin; B25N, B27D, desB30 human insulin; A8H, B25N, B27D, desB30 human insulin; B25H, B27D, desB309 human insulin; A8H, B25H, B27D, desB30 human insulin; A(-1 )P, A(0)P, A14E, B25H, desB30 human insulin; A14E, B(-1 )P, Β(0)Ρ, B25H, desB30 human insulin; A(-1 )P, A(0)P, A14E, B(-1 )P, B(0)P, B25H, desB30 human insulin; A14E, B25H, B30T, B31 L, B32E human insulin; A14E, B25H human insulin; A14E, B16H, B25H, desB30 human insulin; A14E, B10P, B25H, desB30 human insulin; A14E, B10E, B25H, desB30 human insulin; A14E, B4E, B25H, desB30 human insulin; A14H, B16H, B25H, desB30 human insulin; A14H, B10E, B25H, desB30 human insulin; A13H, A14E, B10E, B25H, desB30 human insulin; A13H, A14E, B25H, desB30 human insulin; A14E, A18Q, B3Q, B25H, desB30 human insulin; A14E, B24H, B25H, desB30 human insulin; A14E, B25H, B26G, B27G, B28G, desB30 human insulin; A14E, A21 G, B25H, B26G, B27G, B28G, desB30 human insulin; A14E, A18Q, A21 Q, B3Q, B25H, desB30 human insulin;
A14E, A18Q, A21 Q, B3Q, B25H, B27E, desB30 human insulin; A14E, A18Q, B3Q, B25H, desB30 human insulin; A13H, A14E, B1 E, B25H, desB30 human insulin; A13N, A14E, B25H, desB30 human insulin; A13N, A14E, B1 E, B25H, desB30 human insulin; A(-2)G, A(-1 )P, A(0)P, A14E, B25H, desB30 human insulin; A14E, B(-2)G, B(-1 )P, B(0)P, B25H, desB30 human insulin; A(-2)G, A(-1 )P, A(0)P, A14E, B(-2)G, B(-1 )P, B(0)P, B25H, desB30 human insulin; A14E, B27R, B28D, B29K, desB30 human insulin; A14E, B25H, B27R, B28D, B29K, desB30 human insulin; A14E, B25H, B26T, B27R, B28D, B29K, desB30 human insulin; A14E, B25H, B27R, desB30 human insulin; A14E, B25H, B27H, desB30 human insulin; A14E, A18Q, B3Q, B25H, desB30 human insulin; A13E, A14E, B25H, desB30 human insulin; A12E, A14E, B25H, desB30 human insulin; A15E, A14E, B25H, desB30 human insulin; A13E, B25H, desB30 human insulin; A12E, B25H, desB30 human insulin; A15E, B25H, desB30 human insulin; A14E, B25H, desB27, desB30 human insulin; A14E, desB27, desB30 human insulin; A14H, desB27, desB30 human insulin; A14E, B16H, desB27, desB30 human insulin; A14H, B16H, desB27, desB30 human insulin; A14E, B25H, B26D, B27E, desB30 human insulin; A14E, B25H, B27R, desB30 human insulin; A14E, B25H, B27N, desB30 human insulin; A14E, B25H, B27D, desB30 human insulin; A14E, B25H, B27Q, desB30 human insulin; A14E, B25H, B27E, desB30 human insulin; A14E, B25H, B27G, desB30 human insulin; A14E, B25H, B27H, desB30 human insulin; A14E, B25H, B27K, desB30 human insulin; A14E, B25H, B27P, desB30 human insulin; A14E, B25H, B27S, desB30 human insulin; A14E, B25H, B27T, desB30 human insulin; A13R, A14E, B25H, desB30 human insulin; A13N, A14E, B25H, desB30 human insulin; A13D, A14E, B25H, desB30 human insulin; A13Q, A14E, B25H, desB30 human insulin; A13E, A14E, B25H, desB30 human insulin; A13G, A14E, B25H, desB30 human insulin; A13H, A14E, B25H, desB30 human insulin; A13K, A14E, B25H, desB30 human insulin; A13P, A14E, B25H, desB30 human insulin; A13S, A14E, B25H, desB30 human insulin; A13T, A14E, B25H, desB30 human insulin; A14E, B16R, B25H, desB30 human insulin; A14E, B16D, B25H, desB30 human insulin; A14E, B16Q, B25H, desB30 human insulin; A14E, B16E, B25H, desB30 human insulin; A14E, B16H, B25H, desB30 human insulin; A14R, B25H, desB30 human insulin; A14N, B25H, desB30 human insulin; A14D, B25H, desB30 human insulin; A14Q, B25H, desB30 human insulin; A14E, B25H, desB30 human insulin; A14G, B25H, desB30 human insulin; A14H, B25H, desB30 human insulin; A8H, B10D, B25H human insulin; and A8H, A14E, B10E, B25H, desB30 human insulin and this embodiment may, optionally, comprise B25H, desB30 human insulin and B25N, desB30 human insulin. In a preferred embodiment, a N-terminally modified insulin according to the invention has a peptide part which is selected from the group consisting of: A14E, B25H, desB30 human insulin; A14E, B16H, B25H, desB30 human insulin; A14E, B16E, B25H, desB30 human insulin; A14E, desB27, desB30 human insulin; A14E, B16H, desB27, desB30 human insulin; A14E, B25H, B26G, B27G, B28G, desB30 human insulin; B25H, desB30 human insulin and A14E, B25H, desB27, desB30 human insulin.
In a preferred embodiment, a N-terminally modified insulin according to the invention has a peptide part which is selected from any one of the insulins mentioned above that, in addition, are containing the desB27 mutation.
In a preferred embodiment, a N-terminally modified insulin according to the invention has a peptide part which is selected from the group consisting of: A14E, B25H, desB27, desB30 human insulin; A14E, B16H, B25H, desB27, desB30 human insulin; A14E, desB27, desB30 human insulin; A14E, B16E, B25H, desB27, desB30 human insulin; and B25H, desB27, desB30 human insulin.
In one embodiment, a N-terminally modified insulin according to the invention has a peptide part which is selected from any of the above mentioned insulins and, in addition, comprise one or two of the following mutations in position A21 and/or B3 to improve chemical stability: A21 G, desA21 , B3Q, or B3G.
In a preferred embodiment, a N-terminally modified insulin according to the invention has a peptide part which is selected from the group consisting of: A14E, A21 G, B25H, desB30 human insulin; A14E, A21 G, B16H, B25H, desB30 human insulin; A14E, A21 G, B16E, B25H, desB30 human insulin; A14E, A21 G, B25H, desB27, desB30 human insulin; A14E, A21 G, B25H, desB27, desB30 human insulin; A14E, A21 G, B25H, B26G, B27G, B28G, desB30 human insulin; A21 G, B25H, desB30 human insulin and A21 G, B25N, desB30 human insulin, and, preferably, it is selected from the following protease stabilised insulins: A14E, A21 G, B25H, desB30 human insulin; A14E, A21 G, desB27, desB30 human insulin; A14E, A21 G, B16H, B25H, desB30 human insulin; A14E, A21 G, B16E, B25H, desB30 human insulin; A14E, A21 G, B25H, desB27, desB30 human insulin; A14E, A21 G, B25H, desB27, desB30 human insulin; A21 G, B25H, desB30 human insulin and A21 G, B25N, desB30 human insulin.
Herein, the term "acylated insulin" covers modification of insulin by attachment of one or more lipophilic substituents optionally via a linker to the insulin peptide.
A "lipophilic substituent" is herein understood as a side chain consisting of a fatty acid or a fatty diacid attached to the insulin, optionally via a linker, in an amino acid position such as LysB29, or equivalent. The insulin peptide may be present in an amount of a pharmaceutical composition according to the invention in up to about 20% such as up to about 10% by weight of the total pharmaceutical composition, or from about 0.1 % such as from about 1 %. In one embodiment of the invention, the insulin peptide is present in an amount from about 0.1 % to about 20%, in a further embodiment from about 0.1 % to 15%, 0.1 % to 10%, 1 % to 8% or from about 1 % to 5% by weight of the total composition. It is intended, however, that the choice of a particular level of insulin peptide will be made in accordance with factors well-known in the
pharmaceutical arts, including the solubility of the insulin peptide in the polar organic solvent or optional hydrophilic component or surfactant used, or a mixture thereof, mode of administration and the size and condition of the patient.
Each unit dosage will suitably contain from 1 mg to 200 mg insulin peptide, e.g. about 1 mg, 5 mg, 10 mg, 15 mg, 25 mg, 50 mg, 80 mg, 90 mg, 100 mg, 150 mg, 200 mg insulin peptide, e.g. between 5 mg and 200 mg of insulin peptide. In one embodiment of the invention each unit dosage contains between 10 mg and 200 mg of insulin peptide. In a further embodiment a unit dosage form contains between 10 mg and 100 mg of insulin peptide.
One embodiment of the invention, the unit dosage form contains between 20 mg and 80 mg of insulin peptide. In yet a further embodiment of the invention, the unit dosage form contains between 30 mg and 60 mg of insulin peptide.
In one embodiment of the invention, the unit dosage form contains between 30 mg and 50 mg of insulin peptide. Such unit dosage forms are suitable for administration 1 -5 times daily depending upon the particular purpose of therapy.
The production of polypeptides and peptides such as insulin is well known in the art. Polypeptides or peptides may for instance be produced by classical peptide synthesis, e.g. solid phase peptide synthesis using t-Boc or Fmoc chemistry or other well established techniques, see e.g. Greene and Wuts, "Protective Groups in Organic Synthesis", John Wiley & Sons, 1999. The polypeptides or peptides may also be produced by a method which comprises culturing a host cell containing a DNA sequence encoding the (poly)peptide and capable of expressing the (poly)peptide in a suitable nutrient medium under conditions permitting the expression of the peptide. For (poly)peptides comprising non-natural amino acid residues, the recombinant cell should be modified such that the non-natural amino acids are incorporated into the (poly)peptide, for instance by use of tRNA mutants.
As used herein, the term "microemulsion preconcentrate" means a composition, which spontaneously forms a microemulsion or a nanoemulsion, e.g., an oil-in-water microemulsion or nanoemulsion, swollen micelle, micellar solution, in an aqueous medium, e.g. in water or in the gastrointestinal fluids after oral application. The composition self- emulsifies upon dilution in an aqueous medium for example in a dilution of 1 :5, 1 :10, 1 :50, 1 :100 or higher. In one embodiment the composition according to the present invention forms the microemuslion or nanoemulsion comprising particles or domains of a size below 100nm in diameter. The term "domain size" or "particle size" as used herein refers to repetitive scattering units and may be measured by e.g., small angle X-ray. In one embodiment of the invention, the domain size is smaller than 150nm, in another embodiment, smaller than 100nm and in another embodiment, smaller than 50nm, in another embodiment, smaller than 20nm, in another embodiment, smaller than 15nm, in yet another embodiment, smaller than 10nm.
"SEDDS" (self emulsifying drug delivery systems) are herein defined as mixtures of a hydrophilic component, a surfactant, optionally a co-surfactant or lipid component and a therapeutic macromolecule that forms spontaneously a fine oil in water emulsion when exposed to aqueous media under conditions of gentle agitation or digestive motility that would be encountered in the Gl tract. "SMEDDS" (self micro-emulsifying drug delivery systems) are herein defined as isotropic mixtures of a hydrophilic component a surfactant, optionally a co-surfactant or lipid component and a therapeutic macromolecule that rapidly form an oil in water microemulsion or nanoemulsion when exposed to aqueous media under conditions of gentle agitation or digestive motility that would be encountered in the GI tract. "SNEDDS" (self nano-emulsifying drug delivery systems) are herein defined as isotropic mixtures of a hydrophilic component, at least one surfactant with HLB above 10, optionally a co-surfactant and optionally a lipid component and a therapeutic macromolecule that rapidly form a nanoemulsion (droplet size below 20nm in diameter as e.g. measured by PCS) when exposed to aqueous media under conditions of gentle agitation or digestive motility that would be encountered in the GI tract.
As used herein, the term "emulsion" refers to a slightly opaque, opalescent or opague colloidal coarse dispersion that is formed spontaneously or substantially
spontaneously when its components are brought into contact with an aqueous medium.
As used herein, the term "microemulsion" refers to a clear or translucent, slightly opaque, opalescent, non-opaque or substantially non-opaque colloidal dispersion that is formed spontaneously or substantially spontaneously when its components are brought into contact with an aqueous medium.
A microemulsion is thermodynamically stable and contains homogenously dispersed particles or domains, for example of a solid or liquid state (e.g., liquid lipid particles or droplets), of a mean diameter of less than 150 nm as measured by standard light scattering techniques, e.g., using a MALVERN ZETASIZER Nano ZS. In one embodiment when the pharmaceutical composition according to the invention is brought into contact with an aqueous medium a microemulsion is formed which contains homogenously dispersed particles or domains of a mean diameter of less than 100nm, such as less than 50nm, less than 40nm and less than 30nm. Thus, the term "Z average (nm)" refers to the partice size of the particles or domains of said microemulsion. The term "PDI" is the abbreviation of the term "polydispersity index" and is a measure of the heterogeneity of sizes of molecules or particles in a mixture.
The term "domain size" as used herein refers to repetitive scattering units and may be measured by e.g., small angle X-ray. In one embodiment of the invention, the domain size is smaller than 150 nm, In one embodiment, smaller than 100 nm and In one embodiment, smaller than 50 nm, In one embodiment, smaller than 20 nm, In one embodiment, smaller than 15 nm, in yet another embodiment, smaller than 10 nm.
As used herein, the term "nanoemulsion" refers to a clear or translucent, slightly opaque, opalescent, non-opaque or substantially non-opaque colloidal dispersion with particle or droplet size below 20 nm in diameter (as e.g. measured by PCS) that is formed spontaneously or substantially spontaneously when its components are brought into contact with an aqueous medium. In one embodiment when the pharmaceutical composition according to the invention is brought into contact with an aqueous medium a microemulsion is formed which contains homogenously dispersed particles or domains of a mean diameter of less than 20 nm, such as less than 15 nm, less than 10 nm and greater than about 2-4 nm.
As used herein the term "spontaneously dispersible" when referring to a pre- concentrate refers to a composition that is capable of producing colloidal structures such as nanoemulsions, microemulsions, emulsions and other colloidal systems, when diluted with an aqueous medium when the components of the composition of the invention are brought into contact with an aqueous medium, e.g. by simple shaking by hand for a short period of time, for example for ten seconds. In one embodiment a spontaneously dispersible concentrate according to the invention is a SEDDS, SMEDDS or SNEDDS.
The term "non-ionic surfactant" as used herein refers to any substance, in particular a detergent, that can adsorb at surfaces and interfaces, like liquid to air, liquid to liquid, liquid to container or liquid to any solid and which has no charged groups in its hydrophilic group(s) (sometimes referred to as "heads"). The non-ionic surfactant may be selected from a detergent such as ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglyceri- des and sorbitan fatty acid esters, polysorbate such as polysorbate-20, polysorbate-40, polysorbate-60, polysorbate-80, super refined polysorbate 20, super refined polysorbate 40, super refined polysorbate 60 and super refined polysorbate 80 (where the term "super refined" is used by the supplier Croda for their high purity Tween products), poloxamers such as poloxamer 188 and poloxamer 407, polyoxyethylene sorbitan fatty acid esters,
polyoxyethylene derivatives such as alkylated and alkoxylated derivatives (Tweens, e.g. Tween-20 or Tween-80), block copolymers such as polyethyleneoxide/polypropyleneoxide block copolymers (e.g. Pluronics/Tetronics, Triton X-100 and/or Synperonic PE/L 44 PEL) and ethoxylated sorbitan alkanoates surfactants (e. g. Tween-20, Tween-40, Tween-80, Brij- 35), diglycerol laurate, diglycerol caprate, diglycerol caprylate, diglycerol monocaprylate, polyglycerol laurate, polyglycerol caprate and polyglycerol caprylate.
The term "non-aqueous" as used herein refers to a composition to which no water is added during preparation of the pharmaceutical composition. It is known to the person skilled in the art that a composition which has been prepared without addition of water may take up small amounts of water from the surroundings during handling of the pharmaceutical composition such as e.g. a soft-capsule or a hard-capsule used to encapsulate the composition. Also, the insulin peptide and/or one or more of the excipients in the
pharmaceutical composition may have small amounts of water bound to it before preparing a pharmaceutical composition according to the invention. A non-aqueous pharmaceutical composition according to the invention may thus contain small amounts of water. In one embodiment a non-aqueous pharmaceutical composition according to the invention comprises less than 10%(w/w) water. In another embodiment, the composition according to the invention comprises less than 5%(w/w) water. In another embodiment, the composition according to the invention comprises less than 4%(w/w) water, in another embodiment less than 3%(w/w) water, in another embodiment less than 2%(w/w) water and in yet another embodiment less than 1 %(w/w) water . In one embodiment the composition accord 0%(w/w) water
Examples of other non-ionic surfactants include, but are not limited to:
1 . Reaction products of a natural or hydrogenated castor oil and ethylene oxide. The natural or hydrogenated castor oil may be reacted with ethylene oxide in a molar ratio of from about 1 :35 to about 1 :60, with optional removal of the PEG component from the products. Various such surfactants are commercially available, e.g., the CREMOPHOR series from BASF Corp. (Mt. Olive, NJ), such as CREMOPHOR RH 40 which is PEG40 hydrogenated castor oil which has a saponification value of about 50- to 60, an acid value less than about one, a water content, i.e., Fischer, less than about 2%, an nD 60 of about 1.453-1 .457, and an HLB of about 14-16; 2. Polyoxyethylene fatty acid esters that include polyoxyethylene stearic acid esters, such as the MYRJ series from Uniqema e.g., MYRJ 53 having a m.p. of about 47°C.
Particular compounds in the MYRJ series are, e.g., MYRJ 53 having an m.p. of about 47°C and PEG-40-stearate available as MYRJ 52;
3. Sorbitan derivatives that include the TWEEN series from Uniqema, e.g., TWEEN 60;
4. Polyoxyethylene-polyoxypropylene co-polymers and block co-polymers or poloxamers, e.g., Pluronic F127 or Pluronic F68 from BASF or Synperonic PE/L from Croda;.
5. Polyoxyethylene alkyl ethers, e.g., such as polyoxyethylene glycol ethers of Ci2-Ci8 alcohols, e.g., polyoxyl 10- or 20-cetyl ether or polyoxyl 23-lauryl ether, or 20-oleyl ether, or polyoxyl 10-, 20- or 100-stearyl ether, as known and commercially available as the BRIJ series from Uniqema. Particularly useful products from the BRIJ series are BRIJ 58; BRIJ 76; BRIJ 78; BRIJ 35, i.e. polyoxyl 23 lauryl ether; and BRIJ 98, i.e., polyoxyl 20 oleyl ether. These products have a m.p. between about 32°C to about 43°C;
6. Water-soluble tocopheryl PEG succinic acid esters available from Eastman Chemical Co. with a m.p. of about 36°C, e.g, TPGS, e.g., vitamin E TPGS.
7. PEG sterol ethers having, e.g., from 5-35 [CH2-CH,-0] units, e.g., 20-30 units, e-g., SOLULAN C24 (Choleth-24 and Cetheth-24) from Chemron (Paso Robles, CA); similar products which may also be used are those which are known and commercially available as NIKKOL BPS-30 (polyethoxylated 30 phytosterol) and NIKKOL BPSH-25 (polyethoxylated 25 phytostanol) from Nikko Chemicals;
8. Polyglycerol fatty acid esters, e.g., having a range of glycerol units from 4-10, or 4, 6 or 10 glycerol units. For example, particularly suitable are deca-/hexa-/tetraglyceryl monostearate, e.g., DECAGLYN, HEXAGLYN and TETRAGLYN from Nikko Chemicals;
9. Alkylene polyol ether or ester, e.g., lauroyl macrogol-32 glycerides and/or stearoyl macrogol-32 glycerides which are GELUCIRE 44/14 and GELUCIRE 50/13 respectively;
10. Polyoxyethylene mono esters of a saturated Cio to C22, such as Ci8 substituted e.g.
hydroxy fatty acid; e.g. 12 hydroxy stearic acid PEG ester, e.g. of PEG about e.g. 600-900 e.g. 660 Daltons MW, e.g. SOLUTOL HS 15 from BASF (Ludwigshafen, 20 Germany).
According to a BASF technical leaflet MEF 151 E (1986), SOLUTOL HS 15 comprises about 70% polyethoxylated 12-hydroxystearate by weight and about 30% by weight unesterified polyethylene glycol component. It has a hydrogenation value of 90 to 1 10, a saponification value of 53 to 63, an acid number of maximum 1 , and a maximum water content of 0.5% by weight; 1 1. Polyoxyethylene-polyoxypropylene-alkyl ethers, e.g. polyoxyethylene-polyoxypropylene- ethers of Q2 to C|8 alcohols, e.g. polyoxyethylen-20-polyoxypropylene-4-cetylether which is commercially available as NIKKOL PBC 34 from Nikko Chemicals;
12. Polyethoxylated distearates, e.g. commercially available under the tradenames ATLAS G 1821 from Uniqema and NIKKOCDS-6000P from Nikko Chemicals.
When used herein the term "Hydrophilic-lipophilic balance" or "HLB" of a surfactant or lipophilic component is a measure of the degree to which it is hydrophilic or lipophilic, determined by calculating values for the different regions of the molecule, as described by Griffin (Griffin WC: "Classification of Surface-Active Agents by 'HLB,'" Journal of the Society of Cosmetic Chemists 1 (1949): 31 1 ) or by Davies (Davies JT: "A quantitative kinetic theory of emulsion type, I. Physical chemistry of the emulsifying agent," Gas/Liquid and
Liquid/Liquid Interface. Proceedings of the International Congress of Surface Activity (1957): 426-438).
"Non-ionic surfactants with HLB above 10" are a selection of non-ionic surfactants which have the common feature of having HLB above 10.
For exemplification, a non-limiting list of surfactants with HLB above 10 is provided below together with their HLB value:
Polyethylene glycol sorbitane monolaurate (Tween 20, Polysorbate 20, super refined polysorbate 20) with an HLB of 16.7;
Polyoxyethylene (20) sorbitan monooleate (Tween 80, Polysorbate 80, super refined polysorbate 80) with an HLB of 15;
Polyoxyethylene (20) sorbitan monopalmitate (Tween 40, Polysorbate 40, super refined polysorbate 40) with an HLB of 15.6;
Diglycerol caprylate (diglycerol monocaprylate, polyglycerol caprylate) with an HLB of 1 1 . Polyglycerol caprate (Rylo PG10 Pharma) with HLB of 10;
Caprylocaproyl macrogolglycerides (Labrasol, Labrasol ALF) with an HLB of 14;
Block polymers such as SYNPERONIC PE/L 44 (Poloxamer 124);
Polyoxyethylenestearate (Myrj 45, Macrogolstearate) with HLB of 1 1 .1 ;
Polyoxyethylenestearate (Myrj 49, Macrogolstearate) with HLB of 15;
Polyoxyethylenestearate (Myrj 51 , Macrogolstearate) with HLB of 16;
Polyoxyethylenestearate (Myrj 52, Macrogolstearate) with HLB of 16.9;
Polyoxyethylenestearate (Myrj 53, Macrogolstearate) with HLB of 17.9;
Polyoxyethylenestearate (Myrj 59, Macrogolstearate) with HLB of 18.8; and
Polyoxyethyleneglyceroltriricinoleat (Cremophor EL) with HLB of 13.3. As used herein the term "amino acid" refers to any molecule that contains both amine and carboxyl functional groups.
The term "enteric coating" as used herein means a polymer coating that controls disintegration and release of the solid oral dosage form. The site of disintegration and release of the solid dosage form may be designed depending on the pH of the targeted area, where absorbtion of the therapeutic macromolecule (i.e. therapeutical active peptide or protein) is desired, thus does also include acid resistant protective coatings. The term includes known enteric coatings, but also any other coating with enteric properties, wherein said term "enteric properties" means properties controlling the disintegration and release of the solid oral dosage form (i.e. the oral pharmaceutical composition according to this invention).
The term "enteric soft- or hard capsule technology" when used herein means soft- or hard capsule technology comprising at least one element with enteric properties, such as at least one layer of an enteric coating. The term "delayed release coatings" as used herein means a polymer coating which releases the API in a delayed manner after oral dosing. Delayed release can be achieved by pH dependent or pH independent polymer coatings.
The term "co-surfactant" when used herein refers to an additional surfactant added to a composition or formulation, wherein a first surfactant is present.
In the present context, 1 ,2-propanediol and propylene glycol is used
interchangeably.
THE FOLLOWING IS A NON-LIMITING LIST OF ASPECTS FUTHER COMPRISED WITHIN THE SCOPE OF THE INVENTION:
1 . An oral pharmaceutical composition comprising
a. at least one fatty acid acylated amino acid of the general formula:
Figure imgf000056_0001
wherein R1 is a fatty acid chain comprising 8 to 18 carbon atoms, R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), and R3 is either H, or a salt of, and R4 is a non-cationic amino acid side chain and b. at least one therapeutic macromolecule.
An oral pharmaceutical composition comprising
a. at least one fatty acid acylated amino acid of the general formula:
Figure imgf000057_0001
wherein R1 is a fatty acid chain comprising 8 to 18 carbon atoms,
R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), and
R3 is either H, or a salt thereof, and
R4 is a non-cationic amino acid side chain, and
b. at least one hydrophillic peptide or protein.
An oral pharmaceutical composition comprising
a. at least one fatty acid acylated amino acid of the general formula
Figure imgf000057_0002
wherein R1 is a fatty acid chain comprising 8 to 18 carbon atoms,
R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), and
R3 is either H, or a salt thereof, and
R4 is a non-cationic amino acid side chain, and
b. at least one insulin peptide. An oral pharmaceutical composition according to any of the preceding aspects, wherein the amino acid residue of said at least one fatty acid acylated amino acid is based on a nonpolar hydrophobic amino acid.
An oral pharmaceutical composition according to any of the preceding aspects, wherein the amino acid residue of said at least one fatty acid acylated amino acid is based on a polar uncharged amino acid. An oral pharmaceutical composition according to any of the preceding aspects, wherein the amino acid residue of said at least one fatty acid acylated amino acid is a based on a polar acidic amino acid.
A solid oral composition according to any of the preceeding aspects further comprising at least one insulin.
A solid oral composition according to any of the preceedingaspects further comprising an enteric or delayed release coating.
An oral pharmaceutical composition according to any of the preceeding aspects, wherein the fatty acid moiety of said FA-aa is in the form of its free acid or salt.
An oral pharmaceutical composition according to any of the preceeding aspects wherein said fatty acid moiety of the FA-aa consists of 10 carbon atoms.
An oral pharmaceutical composition according to any of the preceeding aspects, wherein the fatty acid moiety of said FA-aa consists of 12 carbon atoms.
An oral pharmaceutical composition according to any of the preceeding aspects, wherein the fatty acid moiety of said FA-aa consists of 14 carbon atoms.
An oral pharmaceutical composition according to any of the preceeding aspects, wherein the fatty acid moiety of said FA-aa consists of 16 carbon atoms.
An oral pharmaceutical composition according to any of the preceeding aspects, wherein the amino acid residue of said FA-aa is selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and
Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu)..
An oral pharmaceutical composition according to any of the preceeding aspects, wherein the amino acid residue of said FA-aa is selected from the group consisting of the form of the free acid or salt of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
An oral pharmaceutical composition according to any of the preceeding aspects, wherein the FA-aa is selected from the group consisting of: Sodium lauroyl alaninate, N-dodecanoyl-L-alanine, Sodium lauroyl asparaginate, N-dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyl glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium lauroyi glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyi glycinate, N-dodecanoyl-L-glycine, Sodium lauroyi histidinate, N-dodecanoyl-L- histidine, Sodium lauroyi isoleucinate, N-dodecanoyl-L-isoleucine, Sodium lauroyi leucinate, N-dodecanoyl-L-leucine, Sodium lauroyi methioninate, N-dodecanoyl-L- methionine, Sodium lauroyi phenylalaninate, N-dodecanoyl-L-phenylalanine, Sodium lauroyi prolinate, N-dodecanoyl-L-proline, Sodium lauroyi serinate, N-dodecanoyl-L- serine, Sodium lauroyi threoninate, N-dodecanoyl-L-threonine, Sodium lauroyi tryptophanate, N-dodecanoyl-L-tryptophane, Sodium lauroyi tyrosinate, N-dodecanoyl- L-tyrosine, Sodium lauroyi valinate, N-dodecanoyl-L-valine, Sodium lauroyi sarcosinate, N-dodecanoyl-L-sarcosine, Sodium capric alaninate, N-decanoyl-L- alanine, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N-decanoyl-L- cysteine, Sodium capric glutamic acid, N-decanoyl-L-glutamic acid, Sodium capric glutaminate, N-decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric histidinate, N-decanoyl-L-histidine, Sodium capric isoleucinate, N- decanoyl-L-isoleucine, Sodium capric leucinate, N-decanoyl-L-leucine, Sodium
Sodium capric methioninate, N-decanoyl-L-methionine, Sodium capric
phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric prolinate, N-decanoyl-L- proline, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N- decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L-tryptophane,
Sodium capric tyrosinate, N-decanoyl-L-tyrosine, Sodium capric valinate, N-decanoyl- L-valine, Sodium capric sarcosinate and N-decanoyl-L-sarcosine, Sodium lauroyi sarcosinate, Sodium oleoyl sarcosinate, Sodium N-decyl leucine, Amisoft HS-1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyi Glutamate), Amisoft CS-1 1 (Sodium Cocoyl Glutamate),
Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyi sarcosinate, Sodium N- decyl leucine, Sodium cocoyl glycine and Sodium cocoyl glutamate..
An oral pharmaceutical composition according to any of the preceeding aspects, wherein the FA-aa is selected from the group consisting of: Sodium lauroyi alaninate, N-dodecanoyl-L-alanine, Sodium lauroyi asparaginate, N-dodecanoyl-L-asparagine,
Sodium lauroyi aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyi cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyi glutamic acid, N-dodecanoyl-L-glutamic acid, Sodium lauroyi glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyi glycinate, N-dodecanoyl-L-glycine, Sodium lauroyi histidinate, N-dodecanoyl-L- histidine, Sodium lauroyi isoleucinate, N-dodecanoyl-L-isoleucine, Sodium lauroyi leucinate, N-dodecanoyl-L-leucine, Sodium lauroyi methioninate, N-dodecanoyl-L- methionine, Sodium lauroyi phenylalaninate, N-dodecanoyl-L-phenylalanine, Sodium lauroyi prolinate, N-dodecanoyl-L-proline, Sodium lauroyi serinate, N-dodecanoyl-L- serine, Sodium lauroyi threoninate, N-dodecanoyl-L-threonine, Sodium lauroyi tryptophanate, N-dodecanoyl-L-tryptophane, Sodium lauroyi tyrosinate, N-dodecanoyl-
L-tyrosine, Sodium lauroyi valinate, N-dodecanoyl-L-valine, Sodium lauroyi sarcosinate, N-dodecanoyl-L-sarcosine, Sodium capric alaninate, N-decanoyl-L- alanine, Sodium capric asparaginate, N-decanoyl-L-asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N-decanoyl-L- cysteine, Sodium capric glutamic acid, N-decanoyl-L-glutamic acid, Sodium capric glutaminate, N-decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric histidinate, N-decanoyl-L-histidine, Sodium capric isoleucinate, N- decanoyl-L-isoleucine, Sodium capric leucinate, N-decanoyl-L-leucine, Sodium capric methioninate, N-decanoyl-L-methionine, Sodium capric phenylalaninate, N-decanoyl- L-phenylalanine, Sodium capric prolinate, N-decanoyl-L-proline, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L-tryptophane, Sodium capric tyrosinate, N- decanoyl-L-tyrosine, Sodium capric valinate, N-decanoyl-L-valine, Sodium capric sarcosinate and N-decanoyl-L-sarcosine, Sodium lauroyi sarcosinate, Sodium oleoyl sarcosinate, Sodium N-decyl leucine, Amisoft HS-1 1 P (Sodium Stearoyl Glutamate,
Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyi Glutamate), Amisoft CS-1 1 (Sodium Cocoyl Glutamate), Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyi sarcosinate, Sodium N-decyl leucine and Sodium cocoyl glycine, Sodium cocoyl glutamate.
18. An oral pharmaceutical composition according to any of the preceeding aspects, further comprising propylene glycol.
19. An oral pharmaceutical composition according to any of the preceeding aspects, further comprising SEDDS, SMEDDS or SNEDDS.
20. An oral pharmaceutical composition according to any of the preceeding aspects, further comprising other pharmaceutical excipients.
21 . An oral pharmaceutical composition according to any of the preceeding aspects for use as a medicament.
22. An oral pharmaceutical composition according to any of the preceeding aspects for use as a medicament for treatment of Diabetes Mellitus. 23. The pharmaceutical composition according to any of the preceding aspects, wherein said hydrophilic peptide or protein is an insulin peptide.
24. The pharmaceutical composition according to any of the preceding aspects, which comprises less than 10%(w/w) water.
25. The oral pharmaceutical composition according to any of the preceding aspects,
wherein the amino acid residue of said at least one fatty acid acylated amino acid is based on a nonpolar hydrophobic amino acid, a polar uncharged amino acid or polar acidic amino acid.
26. The oral composition according to any of the preceeding aspects further comprising an enteric or delayed release coating.
27. The oral pharmaceutical composition according to any of the preceeding aspects, wherein the fatty acid acylated amino acid is in the form of its free acid or salt.
28. The oral pharmaceutical composition according to any of the preceeding embodiments wherein said fatty acid moiety of the FA-aa consists of 8, 10 or 12.
29. The oral pharmaceutical composition according to any of the preceeding embodiments wherein said fatty acid moiety of the FA-aa consists of 14, 16 or 18 carbon atoms. 30. The oral pharmaceutical composition according to any of the preceeding embodiments wherein said fatty acid moiety of the FA-aa consists of 10, 12, 14, 16 or 18 carbon atoms.
31 . The oral pharmaceutical composition according to any of the preceeding embodiments wherein said fatty acid moiety of the FA-aa consists of 10 or 12.
32. The oral pharmaceutical composition according to any of the preceeding aspects, wherein the amino acid residue of said FA-aa is selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine
(Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr), Asparagine (Apn), and
Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
33. The oral pharmaceutical composition according to any of the preceeding aspects, wherein the fatty acid acylated amino acid is selected from the group consisting of: Sodium lauroyl alaninate, N-dodecanoyl-L-alanine, Sodium lauroyl asparaginate, N- dodecanoyl-L-asparagine, Sodium lauroyl aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyl cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyl glutamic acid, N- dodecanoyl-L-glutamic acid, Sodium lauroyl glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyl glycinate, N-dodecanoyl-L-glycine, Sodium lauroyl histidinate, N- dodecanoyl-L-histidine, Sodium lauroyl isoleucinate, N-dodecanoyl-L-isoleucine, Sodium lauroyi leucinate, N-dodecanoyl-L-leucine, Sodium lauroyi methioninate, N- dodecanoyl-L-methionine, Sodium lauroyi phenylalanine, N-dodecanoyl-L- phenylalanine, Sodium lauroyi prolinate, N-dodecanoyl-L-proline, Sodium lauroyi serinate, N-dodecanoyl-L-serine, Sodium lauroyi threoninate, N-dodecanoyl-L- threonine, Sodium lauroyi tryptophanate, N-dodecanoyl-L-tryptophane, Sodium lauroyi tyrosinate, N-dodecanoyl-L-tyrosine, Sodium lauroyi valinate, N-dodecanoyl-L- valine, Sodium lauroyi sarcosinate, N-dodecanoyl-L-sarcosine, Sodium capric alaninate, N-decanoyl-L-alanine, Sodium capric asparaginate, N-decanoyl-L- asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N-decanoyl-L-cysteine, Sodium capric glutamic acid, N-decanoyl-L- glutamic acid, Sodium capric glutaminate, N-decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric histidinate, N-decanoyl-L-histidine, Sodium capric isoleucinate, N-decanoyl-L-isoleucine, Sodium capric leucinate, N- decanoyl-L-leucine, Sodium Sodium capric methioninate, N-decanoyl-L-methionine, Sodium capric phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric prolinate,
N-decanoyl-L-proline, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L- tryptophane, Sodium capric tyrosinate, N-decanoyl-L-tyrosine, Sodium capric valinate, N-decanoyl-L-valine, Sodium capric sarcosinate and N-decanoyl-L-sarcosine, Sodium lauroyi sarcosinate, Sodium oleoyl sarcosinate, Sodium N-decyl leucine, Amisoft HS-
1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyi Glutamate), Amisoft CS-1 1 (Sodium Cocoyl
Glutamate), Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyi sarcosinate, Sodium N-decyl leucine, Sodium cocoyl glycine, Sodium cocoyl glutamate Sodium lauroyi alaninate, N-dodecanoyl-L-alanine, Sodium lauroyi asparaginate, N- dodecanoyl-L-asparagine, Sodium lauroyi aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyi cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyi glutamic acid, N- dodecanoyl-L-glutamic acid, Sodium lauroyi glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyi glycinate, N-dodecanoyl-L-glycine, Sodium lauroyi histidinate, N- dodecanoyl-L-histidine, Sodium lauroyi isoleucinate, N-dodecanoyl-L-isoleucine,
Sodium lauroyi leucinate, N-dodecanoyl-L-leucine, Sodium lauroyi methioninate, N- dodecanoyl-L-methionine, Sodium lauroyi phenylalaninate, N-dodecanoyl-L- phenylalanine, Sodium lauroyi prolinate, N-dodecanoyl-L-proline, Sodium lauroyi serinate, N-dodecanoyl-L-serine, Sodium lauroyi threoninate, N-dodecanoyl-L- threonine, Sodium lauroyi tryptophanate, N-dodecanoyl-L-tryptophane, Sodium lauroyl tyrosinate, N-dodecanoyl-L-tyrosine, Sodium lauroyl valinate, N-dodecanoyl-L- valine, Sodium lauroyl sarcosinate, N-dodecanoyl-L-sarcosine, Sodium capric alaninate, N-decanoyl-L-alanine, Sodium capric asparaginate, N-decanoyl-L- asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N-decanoyl-L-cysteine, Sodium capric glutamic acid, N-decanoyl-L- glutamic acid, Sodium capric glutaminate, N-decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric histidinate, N-decanoyl-L-histidine, Sodium capric isoleucinate, N-decanoyl-L-isoleucine, Sodium capric leucinate, N- decanoyl-L-leucine, Sodium capric methioninate, N-decanoyl-L-methionine, Sodium capric phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric prolinate, N- decanoyl-L-proline, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L- tryptophane, Sodium capric tyrosinate, N-decanoyl-L-tyrosine, Sodium capric valinate, N-decanoyl-L-valine, Sodium capric sarcosinate and N-decanoyl-L-sarcosine, Sodium lauroyl sarcosinate, Sodium oleoyl sarcosinate, Sodium N-decyl leucine, Amisoft HS- 1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium Cocoyl
Glutamate), Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyl sarcosinate, Sodium N-decyl leucine and Sodium cocoyl glycine, Sodium cocoyl glutamate.
The oral pharmaceutical composition according to any of the preceeding aspects, further comprising propylene glycol.
An oral pharmaceutical composition according to any of the preceeding aspects, further comprising SEDDS, SMEDDS or SNEDDS.
The oral pharmaceutical composition according to any of the preceeding aspects, further comprising other pharmaceutical excipients.
The oral pharmaceutical composition according to any of the preceeding aspects for use as a medicament.
The oral pharmaceutical composition according to any of the preceeding aspects for use as a medicament for treatment of Diabetes Mellitus.
Use of an oral pharmaceutical composition according to any of the preceeding aspects, for increasing the bioavailability of said hydrophilic peptide or protein.
Use of an oral pharmaceutical composition according to any of the preceeding aspects, for increasing the bioavailability of said therapeutic macromolecule.
Use of an oral pharmaceutical composition according to any of the preceeding aspects, for increasing the bioavailability of said therapeutic active peptide. Examples of pharmaceutical compositions comprising insulin derivative and fatty acid acylated amino acids.
Example 1
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of fatty acid acylated amino acids. The composition was injectioned into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 6) and the pharmacokinetic profile was retrieved from the resulting records.
The results are shown in Figure 1 .
Example 2
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of sodium N- capric leucine in concentrations of 10 or 20mg/ml_, respectively. The composition was injectioned into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 4-6) and the pharmacokinetic profiles were retrieved from the resulting records.
The results are shown in Figure 2. Example 3
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of sodium lauroyl sarcosinate (10m/ml_) or sodium N-cocoyl sarcosinate (10mg/ml_), respectively. The formulation (-□-) with N-cocoyl sarcosine contained 50% of the co-solvent propylene glycol. The fatty acid chain distribution in the cocoyl sarcosinate was 1 % C6, 8% C8, 6% C10, 48% C12, 18% C14, 8% C16, 6% C18 saturated and 5% C18 unsaturated.
The resulting compositions was injected into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 6) and the pharmacokinetic profiles.
The results are shown in Figure 3.
Example 4
The insulin derivative A14E, B25H, B29K (N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of increasing amounts (3mg/ml_, 10mg/ml_, 30mg/ml_ and 100mg/L) of sodium lauroyl sarcosinate The resulting compositions were injected into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 6) and the pharmacokinetic profiles were retrieved from the resulting records.
The results are shown in Figure 4. Example 5
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of increasing amounts (3mg/ml_, 10mg/ml_, 30mg/ml_ and 100mg/L) of sodium myristoyl glutamate. The resulting compositions were injected into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 4-6) and the pharmacokinetic profiles were retrieved from the resulting records.
The results are shown in Figure 5.
Example 6
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of 10 mg/mL sodium lauroyl sarcosinate. The composition was injected into colon of anaesthetized overnight fasted Sprague-Dawley rats (n = 6) and the pharmacokinetic profiles were retrieved from the resulting records.
The result is shown in Figure 6.
Example 7
Pharmakokinetic profiles were made of the insulin derivative A14E, B25H,
B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of 10 mg/mL oleoyl sarcosinate or in presence of 10 mg/mL cocoyi sarcosinate and 16.5% of the co-solvent propylene glycol. The fatty acid chain distribution in the cocoyi sarcosinate is 1 % C6, 8% C8, 6% C10, 48% C12, 18% C14, 8% C16, 6% C18 saturated and 5% C18 unsaturated.
The composition was injected into mid-jejunum of anaesthetized overnight fasted Sprague- Dawley rats (n = 6) and the pharmacokinetic profiles were retrieved from the resulting records.
The results are shown in Figure 7. Example 8
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of 10mg/ml_ sodium N-myristoyl-L-glutamate, sodium N-lauroyl-L-glutamate, sodium N-cocoyl-L- glutamate, sodium N-cocoyl glycinate or sodium N-steoryl-L-glutamate, respectively.
The resulting compositions were injected into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n = 4-6) and the pharmacokinetic profiles were retrieved from the resulting records.
The results are shown in Figure 8.
Example 9
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of 10 mg/mL sodium N-capric leucine, sodium, N-capric alanine, sodium N-capric phenylalanine, N-capric isoleucine, N-capric aspart, N-lauroyl leucine or N-myristoyl leucine, respectively. The resulting compositions were injected into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n =5- 6) and the pharmacokinetic profiles were retrieved from the resulting records.
The results are shown in Figure 9.
Example 10
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in propylene glycol in presence of sodium N-capric leucine. The resulting composition was injected into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n =6) and the pharmacokinetic profiles were retrieved from the resulting records.
The result is shown in Figure 10.
Example 1 1
Pharmakokinetic profiles were retrieved from the resulting records related to measurements after peroral dosing of an enteric coated tablet comprising 200 mg of sodium lauroyl sarcosinate, 50 mg of soybean trypsine inhibitor (SBTI) and Eudragit® L30-D55 and Eudragit® NE30D for enteric coating further comprising insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG) and desB30 human insulin (120 nmol/kg) after peroral dosing of an enteric coated tablet comprising 200 mg of sodium lauroyl sarcosinate, 50 mg of soybean trypsine inhibitor (SBTI) and Eudragit® L30-D55 and Eudragit® NE30D for enteric coating to male beagle dogs.
The results are shown in Figure 1 1 as single PK profiles. Example 12
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (60 nmol/kg) dissolved in phosphate buffer (pH 7.4) in presence of 10mg sodium lauroyl leucine, a mixtures of 5 mg/mL sodium lauroyl leucine and 5mg/ml_ capric leucine or 10 mg/mL of the commonly used permeation enhancers salicylate, deoxycholate. The resulting compositions were injected into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n =5- 6) and the pharmacokinetic profiles were calculated based on the resulting records.
The result is shown in Figure 12. Example of Liquid non-aqueous pharmaceutical compositions comprising insulin derivative and fatty acid acylated amino acids
Example 13
Liquid insulin SEDDS, SMEDDS and SNEDDS formulations were prepared according to the guidance given in WO08145728 comprising the fatty acid acylated amino acid sodium N- lauroyl phenylalanine.
All formulations contained the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl- gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg).
The insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg) dissolved in liquid SEDDS, SMEDDS and SNEDDS formulations comprising sodium N-lauroyl phenylalanine.
The resulting compositions were injected into mid-jejunum of anaesthetized overnight fasted Sprague-Dawley rats (n =5- 6) and the pharmacokinetic profiles were calculated based on the resulting records.
The compositions are shown in table 1 and PK results are shown in Figure 13.
The compositions are shown in Table 1.
Table 1 Liquid insulin SEDDS, SMEDDS and SNEDDS formulations comprising the co- surfactant sodium N-lauroyl phenylalanine. All formulations contain the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl- gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg).
Figure imgf000068_0001
Table 1 Example 14
Insulin SEDDS and SMEDDS compositions were prepared according to the guidance given in WO08145728 comprising at least one fatty acid acylated amino acid (FA-aa). Mean particle size (hydrodynamic diameter) was analysed after 100 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index). All formulations contained the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (30 nmol/kg).
The results are shown in Table 2.
Table 2 Insulin SEDDS and SMEDDS compositions comprising at least one FA-aa.
Mean particle size (hydrodynamic diameter) is shown after 100 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index).
Mean
Insulin Propylene FA-aa (50 Tween Diglycerol
particle size PDI derivative glycol mg) 20 caprylate
(diameter)
5 mg 150 mg Sodium N- 300 mg 500 mg 170.2 nm 0.226 capric serine
5 mg 150 mg Sodium N- 300 mg 500 mg 75.7 nm 0.131 capric
histidine
5 mg 150 mg Sodium N- 300 mg 500 mg 91.4 nm 0.168 capric
threonine
5 mg 150 mg Sodium N- 300 mg 500 mg 151.1 nm (74%) 0.483 capric 32.0 nm (27%) phenylalanine
5 mg 150 mg Sodium N- 300 mg 500 mg 104.1 nm 0148 capric Mean
Insulin Propylene FA-aa (50 Tween Diglycerol
particle size PDI derivative glycol mg) 20 caprylate
(diameter)
aspartate
5 mg 150 mg Sodium N- 300 mg 500 mg 1 10.6 nm 0.188 capric proline
5 mg 150 mg Sodium N- 300 mg 500 mg 209.1 nm 0.461 capric leucine
5 mg 150 mg Sodium N- 300 mg 500 mg 53.8 nm (75%) 0.442 capric 328 nm (25%) isoleucine
5 mg 150 mg Sodium N- 300 mg 500 mg 71.4 nm 0.130 capric alanine
5 mg 150 mg Sodium N- 300 mg 500 mg 43.1 nm 0.325 capric
tyrosine
5 mg 150 mg Sodium N- 300 mg 500 mg 93.4 nm (80%) 0.482 capric 1092 nm (20%) glutamine
5 mg 150 mg Sodium N- 300 mg 500 mg 61.46 nm 0.157 lauroyl valine
5 mg 150 mg Sodium N- 300 mg 500 mg 30.33 nm (72%) 0.777 lauroyl 867 nm (18%) isoleucine
5 mg 150 mg Sodium N- 300 mg 500 mg 47.04 nm (86%) 0.502 lauroyl 15.23 nm (14%) tyrosine
5 mg 150 mg Sodium N- 300 mg 500 mg 1 19,8 nm 0.162 lauroyl serine
5 mg 150 mg Sodium N- 300 mg 500 mg 61.84 nm 0.198 lauroyl
glycine
5 mg 150 mg Sodium N- 300 mg 500 mg 31.32 nm (87%) 0.272 lauroyl 06.36 nm (13%) trypsine
5 mg 150 mg Sodium N- 300 mg 500 mg 91.07 nm 0.175 lauroyl
alanine
5 mg 150 mg Sodium N- 300 mg 500 mg 87.95 nm 0.185 lauroyl
histidine
5 mg 150 mg Sodium N- 300 mg 500 mg 74.55 nm (62 %) 0.442 lauroyl 686.6 nm (38%) glutamine
5 mg 150 mg Sodium N- 300 mg 500 mg 45.57 nm 0.241 lauroyl
aspartate
5 mg 150 mg Sodium N- 300 mg 500 mg 102.2 nm 0.171 lauroyl proline
Table 2
Example 15 Enteric softcapsule comprising insulin derivative and fatty acid acylated amino acids formulated in a SEDDS. Insulin SEDDS compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa).
Pharmakokinetic profile in a single beagle dog is shown of the insulin derivative A1 (N,N- Dimethyl), A14E, B1 (N, N-dimethyl), B25H, B29K(N(eps)octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (120 nmol/kg) after peroral dosing of an enteric coated soft capsule comprising 30 mg of sodium lauroyi leucine sodium salt, 150 mg of propylene glycol, 300 mg of Polysorbate 20 and 520 mg of diglycerol monocaprylate.
Softcapsule was enteric coated with a mixture of Eudragit® L30-D55 and Eudragit® NE30D. The result is shown in Figure 14 as single PK profile. Example 16
Liquid non-aqueous insulin analogue compositions with different amounts of N-lauroyl leucine sodium salt.Jnsulin SEDDS compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa). Insulin SEDDS and SMEDDS compositions were prepared comprising increasing amounts of N-lauroyl leucine sodium salt. Mean particle size (hydrodynamic diameter) was analysed after 50 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index). All formulations contained the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG- OEG), desB30 human insulin (5mg/g).
The results are shown in Table 3.
Table 3. Liquid insulin analogue compositions comprising different amounts of N-lauroyl leucine sodium salt.
Particle size
Diglycerol (50 fold dilution in MilliQ
Propylen Polysorbat N-lauroyl leucine
NO mono water)
e glycol e 20 sodium salt
caprylate
Z average (nm) PDI Particle size
Diglycerol (50 fold dilution in MilliQ
Propylen Polysorbat N-lauroyl leucine
NO mono water)
e glycol e 20 sodium salt
caprylate
Z average (nm) PDI
1 15% 30% 52% 3% 49 0,126
2 15% 30% 50% 5% 66 0,2
3 15% 30% 48% 7% 1866 1
4 15% 30% 40% 10% 282 0,6
Table 3
Example 17
Liquid insulin analogue compositions with different amounts of N-lauroyl leucine sodium salt further comprising diglycerol monocaprylate and propylene glycol. Insulin SEDDS
compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa). nsulin SEDDS compositions were prepared comprising different amounts of N-lauroyl leucine sodium salt. Mean particle size
(hydrodynamic diameter) was analysed after 50 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index). All formulations contained the insulin derivative A14E, B25H, B29K(N(eps)Octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin (5mg/g).
The results are shown in Table 4.
Table 4. Liquid insulin analogue compositions with different amounts of N-lauroyl leucine sodium salt just comprising diglycerol monocaprylate and propylene glycol.
N-lauroyl Particle size (1:10 dilution mQ
Propylene Diglycerol
leucine water)
No glycol mono
sodium
% caprylate % Z average (nm) PDI
salt %
1 15% 80 5 7.3 0,226 N-lauroyl Particle size (1:10 dilution mQ
Propylene Diglycerol
leucine water)
No glycol mono
sodium
% caprylate %
salt % Z average (nm) PDI
2 15% 77,5 7,5 5.5 0,23
3 15% 75 10 223 0,255
4 15% 72,5 12,5 44 0,281
5 15% 70 15 309 0,384
6 15% 67,5 17,5 330 0,35
7 15% 65 20 769 0,699
8 15% 62,5 22,5 728 0,607
9 15% 60 25 642 0,629
10 15% 57,5 27,5 352 0,359
Table 4
Example 18
Lipid compositions with different fatty acid acylated aminoacids, various solvents and different Polysorbate. Insulin SEDDS compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa).. Insulin SEDDS and SMEDDS compositions were prepared comprising different fatty acid acylated aminoacid sodium salts, polysorbates and solvents. Mean particle size (hydrodynamic diameter) was analysed after 50 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index). All formulations comprise 5 mg/g insulin analogue A1 (N,N-Dimethyl), A14E, B1 (N, N- dimethyl), B25H, B29K(N(eps)octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin 5mg/g. The results are shown in Table 5.
Table 5. Lipid compositions with different fatty acid acylated aminoacids, various solvents and different Polysorbates.
Particle size (1:50
Fatty acid Diglycerol MilliQ water)
Solvent acylated Surfactant mono
No
15% aminoacid 35% caprylate Z average
5% (45%) PDI
(nm)
N-lauroyl
Diglycerol
Propylene leucine
1 Tween 20 mono 570 0.52
glycol sodium
caprylate
salt
N-lauroyl
Diglycerol
Propylene leucine
2 Tween 40 mono 268 0.35
glycol sodium
caprylate
salt
N-lauroyl
Diglycerol
Propylene leucine
3 Tween 60 mono 97 0.128 glycol sodium
caprylate
salt
N-lauroyl
Diglycerol
Propylene leucine
4 Tween 80 mono 115 0.185 glycol sodium
caprylate
salt
N-capric
Diglycerol
Propylene leucine
5 Tween 20 mono 74 0.53
glycol sodium
caprylate
salt
N-capric
Diglycerol
Propylene leucine
6 Tween 40 mono 87 0.56
glycol sodium
caprylate
salt
N-capric
Diglycerol
Propylene leucine
7 Tween 60 mono 12 0.29
glycol sodium
caprylate
salt
N-capric
Diglycerol
Propylene leucine
8 Tween 80 mono 125 0.33
glycol sodium
caprylate
salt
Diglycerol
Propylene N-lauroyl
9 Tween 20 mono 7,5 0.24
glycol sarcosinate
caprylate Particle size (1:50
Fatty acid Diglycerol MilliQ water)
Solvent acylated Surfactant mono
No
15% aminoacid 35% caprylate Z average
5% (45%) PDI
(nm)
Diglycerol
Propylene N-lauroyl
10 Tween 40 mono 7,5 0.26
glycol sarcosinate
caprylate
Diglycerol
Propylene N-lauroyl
11 Tween 60 mono 7,4 0.27
glycol sarcosinate
caprylate
Diglycerol
Propylene N-lauroyl
12 Tween 80 mono 14,7 0.41
glycol sarcosinate
caprylate
N-lauroyl
Diglycerol
leucine
13 H20 Tween 20 mono 178 0.25
sodium
caprylate
salt
N-lauroyl
Diglycerol
leucine
14 H20 Tween 40 mono 740 0.55
sodium
caprylate
salt
N-lauroyl
Diglycerol
leucine
15 H20 Tween 60 mono 196 0.29
sodium
caprylate
salt
N-lauroyl
Diglycerol
leucine
16 H20 Tween 80 mono 170 0.26
sodium
caprylate
salt
Diglycerol
N-lauroyl
17 H20 Tween 20 mono 8 0.27
sarcosinate
caprylate
Diglycerol
N-lauroyl
18 H20 Tween 40 mono 8 0.27
sarcosinate
caprylate
Diglycerol
N-lauroyl
19 H20 Tween 60 mono 10 0.34
sarcosinate
caprylate
Diglycerol
N-lauroyl
20 H20 Tween 80 mono 17.7 0.47
sarcosinate
caprylate
Table 5
Example 19
Liquid lipid based formulations comprising at least one fatty acid acylated aminoacid, insulin derivative, solvent and at least one lipid or co-surfactant were prepared. Insulin SEDDS compositions were prepared according to the guidance given in WO08145728 (in short, the insulin was first dissolved in water and the pH adjusted to pH 7.4 with a non volatile base (NaOH) followed by freeze drying, the resulting insulin powder was then dissolved first in propylene glycol and then mixed with the other excipients as described) comprising at least one fatty acid acylated amino acid (FA-aa).
Insulin SEDDS compositions were prepared comprising different fatty acid acylated aminoacid sodium salts, lipid or co-surfactant and a solvent. Mean particle size
(hydrodynamic diameter) was analysed after 50 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index). All formulations comprise 5 mg/g insulin analogue A1(N,N-Dimethyl), A14E, B1(N, N-dimethyl), B25H, B29K(N(eps)octadecanedioyl-gGlu- OEG-OEG), desB30 human insulin. The results are shown in Table 6.
Table 6. Liquid lipid based formulations comprising at least one fatty acid acylated aminoacid, insulin derivative, solvent and at least one lipid or co-surfactant are described.
Figure imgf000075_0001
Table 6
Example 20
Lipid SEDDS, SMEDDS and SNEDDS compositions comprising N-lauroyl leucine sodium salt and different surfactants with variable HLB values were prepared. Insulin SEDDS compositions were prepared according to the guidance given in WO08145728 comprising at least one fatty acid acylated amino acid (FA-aa). Insulin SEDDS and SMEDDS compositions were prepared comprising N-lauroyl leucine sodium salt, propylene glycol, diglycerol mono caprylate and a high or low HLB surfactant. All formulations comprise 5 mg/g insulin analogue A1(N,N-Dimethyl), A14E, B1(N, N- dimethyl), B25H, B29K(N(eps)octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin.
Mean particle size (hydrodynamic diameter) was analysed after 50 fold dilution in MilliQ water at 37°C and respective PDI (poly dispersity index). All formulations comprise 5 mg/g insulin analogue A1(N,N-Dimethyl), A14E, B1(N, N-dimethyl), B25H,
B29K(N(eps)octadecanedioyl-gGlu-OEG-OEG), desB30 human insulin. The results are shown in Table 7. e 7 were composed as follows;
Figure imgf000076_0001
Table 7. Lipid compositions comprising N-lauroyl leucine sodium salt and different
surfactants with variable HLB values.
Visual
Surfactant (Z)
Formulation appearance
NO or co- HLB average PDI
appearance after
surfactant (nm)
dilution
clear
1 Span 80 4.3 clear liquid 35 0.116
solution
2 Span 60 4.8 insoluble X X X clear liquid turbid
3 Span 40 6.7 2767 0.36
with heating solution clear
4 Span 20 8.6 clear liquid 48 0.44
solution
5 Span 10 9.4 insoluble X X X
clear
6 Span 8 10.3 little turbid 4048 1.0
solution
clear
7 Span 6 11.4 clear liquid 5436 1.0
solution
clear
8 Tween 81 10 clear liquid 60 0.18
solution Visual
Surfactant (Z)
Formulation appearance
NO or co- HLB average PDI appearance after
surfactant (nm)
dilution clear liquid clear
9 Tween 65 10.5 6 0.294 with heating solution clear
10 Tween 85 11 clear liquid 12 0.37 solution
clear
11 Tween 21 13.3 clear liquid 93 0.22 solution
clear
12 Tween 60 14.8 clear liquid 1.7 0.16 solution
clear
13 Tween 80 15 clear liquid 330 0.56 solution
clear
14 Tween 40 15.6 clear liquid 615 0.83 solution
clear
15 Tween 20 16.7 clear liquid 751 1.0 solution
dec- clear
16 Poloxamerl24 clear liquid 185 0.27
18 solution
Deoxycholate clear
17 16 clear liquid 3012 1.0
Na solution
Taurocholate clear
18 N/A clear liquid 1673 1.0
Na solution
Table 7
Example of other compositions
Example 21
The composition of the insulin degludec/liraglutide drug product that Novo Nordisk A/S currently has in clinical development is shown below. This formulation has been shown to be a stable combination product suitable for use in type II diabetes clinical trials (subcutaneous injection)."
Names of ingredients in the drug product formulation
Drug substances
Liraglutide, 3.6 mg (960 nmol) per ml
Insulin degludec, 600 nmol (100 U) per ml
Excipients
Phenol
· Glycerol
Zinc
Formulation process specialities
Both insulin degludec and liraglutide drug substances are added in the form of a solid powder, separately and directly to a mixture of excipients.
All of the zinc is added in one step.
There is no need for holding time anywhere in the formulation process.
While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those of ordinary skill in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.

Claims

1 . An oral pharmaceutical composition comprising
a. at least one fatty acid acylated amino acid of the general formula:
O
4 I 1
R R
wherein R1 is a fatty acid chain comprising 8 to 18 carbon atoms,
R2 is either H (i.e. hydrogen) or CH3 (i.e. methyl group), and
R3 is either H, or a salt thereof, and
R4 is a non-cationic amino acid side chain, and
b. at least one hydrophillic peptide or protein.
2. The pharmaceutical composition according to claim 1 , wherein said hydrophilic
peptide or protein is an insulin peptide.
3. The pharmaceutical composition according to claim 1 , which comprises less than
10%(w/w) water.
4. The oral pharmaceutical composition according to claim 1 , wherein the amino acid residue of said at least one fatty acid acylated amino acid is based on a nonpolar hydrophobic amino acid, a polar uncharged amino acid or polar acidic amino acid.
5. The oral composition according to any of the preceeding claims further comprising an enteric or delayed release coating.
6. The oral pharmaceutical composition according to any of the preceeding claims,
wherein the fatty acid acylated amino acid is in the form of its free acid or salt.
7. The oral pharmaceutical composition according to any of the preceeding embodiments wherein said fatty acid moiety of the fatty acid aclated amino acid consists of 10, 12, 14, 16 or 18 carbon atoms.
8. The oral pharmaceutical composition according to any of the preceeding claims,
wherein the amino acid residue of said fatty acid aclated amino acid is selected from the group consisting of Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (lie), Phenylalanine (Phe), Tryptophane (Trp), Metheonine (Met), Proline (Pro), Sarcosinate, Glycine (Gly), Serine (Ser), Threonine (Thr), Cysteine (Cys), Tyrosine (Tyr),
Asparagine (Apn), and Glutamine (Gin), Aspartic acid (Asp) and Glutamic acid (Glu).
9. The oral pharmaceutical composition according to any of the preceeding claims,
wherein the fatty acid acylated amino acid is selected from the group consisting of: Sodium lauroyi alaninate, N-dodecanoyl-L-alanine, Sodium lauroyi asparaginate, N- dodecanoyl-L-asparagine, Sodium lauroyi aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyi cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyi glutamic acid, N- dodecanoyl-L-glutamic acid, Sodium lauroyi glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyi glycinate, N-dodecanoyl-L-glycine, Sodium lauroyi histidinate, N- dodecanoyl-L-histidine, Sodium lauroyi isoleucinate, N-dodecanoyl-L-isoleucine, Sodium lauroyi leucinate, N-dodecanoyl-L-leucine, Sodium lauroyi methioninate, N- dodecanoyl-L-methionine, Sodium lauroyi phenylalaninate, N-dodecanoyl-L- phenylalanine, Sodium lauroyi prolinate, N-dodecanoyl-L-proline, Sodium lauroyi serinate, N-dodecanoyl-L-serine, Sodium lauroyi threoninate, N-dodecanoyl-L- threonine, Sodium lauroyi tryptophanate, N-dodecanoyl-L-tryptophane, Sodium lauroyi tyrosinate, N-dodecanoyl-L-tyrosine, Sodium lauroyi valinate, N-dodecanoyl-L- valine, Sodium lauroyi sarcosinate, N-dodecanoyl-L-sarcosine, Sodium capric alaninate, N-decanoyl-L-alanine, Sodium capric asparaginate, N-decanoyl-L- asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N-decanoyl-L-cysteine, Sodium capric glutamic acid, N-decanoyl-L- glutamic acid, Sodium capric glutaminate, N-decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric histidinate, N-decanoyl-L-histidine, Sodium capric isoleucinate, N-decanoyl-L-isoleucine, Sodium capric leucinate, N- decanoyl-L-leucine, Sodium Sodium capric methioninate, N-decanoyl-L-methionine,
Sodium capric phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric prolinate, N-decanoyl-L-proline, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L- tryptophane, Sodium capric tyrosinate, N-decanoyl-L-tyrosine, Sodium capric valinate, N-decanoyl-L-valine, Sodium capric sarcosinate and N-decanoyl-L-sarcosine, Sodium lauroyi sarcosinate, Sodium oleoyl sarcosinate, Sodium N-decyl leucine, Amisoft HS- 1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyi Glutamate), Amisoft CS-1 1 (Sodium Cocoyl
Glutamate), Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyi sarcosinate, Sodium N-decyl leucine, Sodium cocoyl glycine, Sodium cocoyl glutamate Sodium lauroyi alaninate, N-dodecanoyl-L-alanine, Sodium lauroyi asparaginate, N- dodecanoyl-L-asparagine, Sodium lauroyi aspartic acid, N-dodecanoyl-L-aspartic acid, Sodium lauroyi cysteinate, N-dodecanoyl-L-cysteine, Sodium lauroyi glutamic acid, N- dodecanoyl-L-glutamic acid, Sodium lauroyi glutaminate, N-dodecanoyl-L-glutamine, Sodium lauroyi glycinate, N-dodecanoyl-L-glycine, Sodium lauroyi histidinate, N- dodecanoyl-L-histidine, Sodium lauroyl isoleucinate, N-dodecanoyl-L-isoleucine, Sodium lauroyl leucinate, N-dodecanoyl-L-leucine, Sodium lauroyl methioninate, N- dodecanoyl-L-methionine, Sodium lauroyl phenylalaninate, N-dodecanoyl-L- phenylalanine, Sodium lauroyl prolinate, N-dodecanoyl-L-proline, Sodium lauroyl serinate, N-dodecanoyl-L-serine, Sodium lauroyl threoninate, N-dodecanoyl-L- threonine, Sodium lauroyl tryptophanate, N-dodecanoyl-L-tryptophane, Sodium lauroyl tyrosinate, N-dodecanoyl-L-tyrosine, Sodium lauroyl valinate, N-dodecanoyl-L- valine, Sodium lauroyl sarcosinate, N-dodecanoyl-L-sarcosine, Sodium capric alaninate, N-decanoyl-L-alanine, Sodium capric asparaginate, N-decanoyl-L- asparagine, Sodium capric aspartic acid, N-decanoyl-L-aspartic acid, Sodium capric cysteinate, N-decanoyl-L-cysteine, Sodium capric glutamic acid, N-decanoyl-L- glutamic acid, Sodium capric glutaminate, N-decanoyl-L-glutamine, Sodium capric glycinate, N-decanoyl-L-glycine, Sodium capric histidinate, N-decanoyl-L-histidine, Sodium capric isoleucinate, N-decanoyl-L-isoleucine, Sodium capric leucinate, N- decanoyl-L-leucine, Sodium capric methioninate, N-decanoyl-L-methionine, Sodium capric phenylalaninate, N-decanoyl-L-phenylalanine, Sodium capric prolinate, N- decanoyl-L-proline, Sodium capric serinate, N-decanoyl-L-serine, Sodium capric threoninate, N-decanoyl-L-threonine, Sodium capric tryptophanate, N-decanoyl-L- tryptophane, Sodium capric tyrosinate, N-decanoyl-L-tyrosine, Sodium capric valinate, N-decanoyl-L-valine, Sodium capric sarcosinate and N-decanoyl-L-sarcosine, Sodium lauroyl sarcosinate, Sodium oleoyl sarcosinate, Sodium N-decyl leucine, Amisoft HS- 1 1 P (Sodium Stearoyl Glutamate, Amisoft MS-1 1 (Sodium Myristoyl Glutamate)), Amisoft LS-1 1 (Sodium Lauroyl Glutamate), Amisoft CS-1 1 (Sodium Cocoyl
Glutamate), Amilite GCS-1 1 (Sodium Cocoyl Glycinate), Sodium lauroyl sarcosinate, Sodium N-decyl leucine and Sodium cocoyl glycine, Sodium cocoyl glutamate.
10. The oral pharmaceutical composition according to any of the preceeding claims,
further comprising propylene glycol.
1 1 . An oral pharmaceutical composition according to any of the preceeding claims, further comprising SEDDS, SMEDDS or SNEDDS.
12. The oral pharmaceutical composition according to any of the preceeding claims, further comprising other pharmaceutical excipients.
13. The oral pharmaceutical composition according to any of the preceeding claims for use as a medicament.
14. The oral pharmaceutical composition according to any of the preceeding claims for use as a medicament for treatment of Diabetes Mellitus.
15. Use of an oral pharmaceutical composition according to any of the preceeding claims, for increasing the bioavailability of said hydrophilic peptide or protein.
PCT/EP2012/056708 2011-04-14 2012-04-12 Fatty acid acylated amino acids for oral peptide delivery WO2012140155A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP12713173.8A EP2696847A1 (en) 2011-04-14 2012-04-12 Fatty acid acylated amino acids for oral peptide delivery
US14/111,274 US20140056953A1 (en) 2011-04-14 2012-04-12 Fatty acid acylated amino acids for oral peptide delivery
CN201280017853.5A CN103458873B (en) 2011-04-14 2012-04-12 For the aminoacid of the fatty-acylation of oral peptide delivery
JP2014504327A JP6030630B2 (en) 2011-04-14 2012-04-12 Fatty acylated amino acids for oral peptide delivery

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP11162411.0 2011-04-14
EP11162411 2011-04-14
US201161477719P 2011-04-21 2011-04-21
US61/477,719 2011-04-21

Publications (1)

Publication Number Publication Date
WO2012140155A1 true WO2012140155A1 (en) 2012-10-18

Family

ID=44343059

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/056708 WO2012140155A1 (en) 2011-04-14 2012-04-12 Fatty acid acylated amino acids for oral peptide delivery

Country Status (5)

Country Link
US (1) US20140056953A1 (en)
EP (1) EP2696847A1 (en)
JP (1) JP6030630B2 (en)
CN (1) CN103458873B (en)
WO (1) WO2012140155A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014060472A1 (en) * 2012-10-17 2014-04-24 Novo Nordisk A/S Fatty acid acylated amino acids for oral peptide delivery
WO2014060512A1 (en) * 2012-10-17 2014-04-24 Novo Nordisk Health Care Ag Fatty acid acylated amino acids for growth hormone delivery
WO2015162195A1 (en) * 2014-04-23 2015-10-29 Novo Nordisk A/S Fatty acid acylated amino acids for oral peptide delivery
EP4180060A1 (en) 2021-11-15 2023-05-17 Adocia Solid compositions comprising a peptide or a protein and an acylated amino acid
WO2023084118A1 (en) 2021-11-15 2023-05-19 Adocia Solid compositions comprising a peptide or a protein and an acylated amino acid
EP4299057A1 (en) 2022-06-30 2024-01-03 Adocia Solid compositions comprising a peptide or a protein and an acylated amino acid
EP4299071A1 (en) 2022-07-01 2024-01-03 Adocia Compositions comprising a peptide or a protein and an acylated amino acid

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6285447B2 (en) * 2012-10-17 2018-02-28 ノヴォ ノルディスク アー/エス Fatty acylated D-amino acids for oral peptide delivery
EP3006045B3 (en) 2014-10-07 2021-03-17 Cyprumed GmbH Pharmaceutical formulations for the oral delivery of peptide or protein drugs
CN104961687B (en) * 2015-06-03 2017-07-25 苏州维泰生物技术有限公司 1,2 diazine derivatives and its preparation, purposes
CN106551902B (en) * 2015-09-17 2020-07-03 阿赖耶识(上海)生物技术有限公司 High-stability non-vesicular nanoparticles and application thereof in treating microbial infection
CA2997343A1 (en) 2015-10-07 2017-04-13 Cyprumed Gmbh Pharmaceutical formulations for the oral delivery of peptide drugs
WO2018065634A1 (en) 2016-10-07 2018-04-12 Cyprumed Gmbh Pharmaceutical compositions for the nasal delivery of peptide or protein drugs
TWI751379B (en) 2017-09-21 2022-01-01 奧孟亞股份有限公司 Pharmaceutical composition for delivery of peptide
US11365177B2 (en) * 2017-11-30 2022-06-21 Dana-Farber Cancer Institute, Inc. Chemical uncouplers of respiration and methods of use thereof
FR3075197B1 (en) * 2017-12-20 2019-11-15 Societe D'exploitation De Produits Pour Les Industries Chimiques Seppic NOVEL COMPOSITION OF LIPOAMINOACIDES AND DIOLS, PROCESS FOR THEIR PREPARATION AND COSMETIC OR PHARMACEUTICAL COMPOSITION RESULTING THEREFROM
US20210087250A1 (en) 2018-04-06 2021-03-25 Cyprumed Gmbh Pharmaceutical compositions for the transmucosal delivery of therapeutic peptides and proteins
US20240041983A1 (en) 2020-09-07 2024-02-08 Cyprumed Gmbh Improved pharmaceutical formulations of glp-1 receptor agonists
WO2023166179A1 (en) 2022-03-03 2023-09-07 Cyprumed Gmbh Improved oral pharmaceutical formulations of therapeutic peptides and proteins

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001035998A1 (en) 1999-11-12 2001-05-25 Pharmaderm Laboratories, Ltd. Compositions for transdermal and transmucosal administration of therapeutic agents
WO2003030865A1 (en) 2001-10-11 2003-04-17 Imi Biomed, Inc. Pro-micelle pharmaceutical compositions
US20040147578A1 (en) 2001-03-28 2004-07-29 Nicolas Calvet Use of lipoaminoacids as absorption promoters in a pharmaceutical composition
WO2004064758A2 (en) 2003-01-21 2004-08-05 Unigene Laboratories Inc. Improved oral delivery of peptides
WO2005012347A2 (en) 2003-08-05 2005-02-10 Novo Nordisk A/S Novel insulin derivatives
US20050282756A1 (en) 2004-06-18 2005-12-22 Mehta Nozer M Oral delivery of peptide pharmaceutical compositions
WO2008034881A1 (en) 2006-09-22 2008-03-27 Novo Nordisk A/S Protease resistant insulin analogues
GB2445013A (en) * 2006-11-29 2008-06-25 Malvern Cosmeceutics Ltd Novel compositions for the solubilisation of hydrophobic substances
WO2008145728A1 (en) 2007-06-01 2008-12-04 Novo Nordisk A/S Spontaneously dispersible preconcentrates including a peptide drug in a solid or semisolid carrier
WO2009115469A1 (en) 2008-03-18 2009-09-24 Novo Nordisk A/S Protease stabilized, acylated insulin analogues
WO2010060667A1 (en) * 2008-11-28 2010-06-03 Novo Nordisk A/S Pharmaceutical compositions suitable for oral administration of derivatized insulin peptides
WO2011068019A1 (en) 2009-12-02 2011-06-09 独立行政法人科学技術振興機構 Retrograde transport viral vector system having envelope comprising fused glycoprotein

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2772168A (en) * 1951-11-28 1956-11-27 Colgate Palmolive Co Fermentable carbohydrate food products
US4258029A (en) * 1979-04-23 1981-03-24 Connaught Laboratories Limited Synthetic adjuvants for stimulation of antigenic responses
US4749694A (en) * 1984-04-26 1988-06-07 Merck & Co., Inc. Novel lysine esters used as absorption
US5270032A (en) * 1990-10-04 1993-12-14 The Research Foundation Of State University Of New York Composition and method for the prevention and treatment of candidiasis
US5498420A (en) * 1991-04-12 1996-03-12 Merz & Co. Gmbh & Co. Stable small particle liposome preparations, their production and use in topical cosmetic, and pharmaceutical compositions
US6624754B1 (en) * 1998-01-20 2003-09-23 Hoffman Resources Llc Personal security and tracking system
FR2767064B1 (en) * 1997-08-07 1999-11-12 Centre Nat Rech Scient METHOD FOR RELEASING AN ACTIVE INGREDIENT CONTAINED IN A MULTIPLE EMULSION
JP4147235B2 (en) * 2004-09-27 2008-09-10 キヤノン株式会社 Discharge liquid, discharge method, droplet forming method, liquid discharge cartridge, and discharge apparatus

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001035998A1 (en) 1999-11-12 2001-05-25 Pharmaderm Laboratories, Ltd. Compositions for transdermal and transmucosal administration of therapeutic agents
US20040147578A1 (en) 2001-03-28 2004-07-29 Nicolas Calvet Use of lipoaminoacids as absorption promoters in a pharmaceutical composition
WO2003030865A1 (en) 2001-10-11 2003-04-17 Imi Biomed, Inc. Pro-micelle pharmaceutical compositions
WO2004064758A2 (en) 2003-01-21 2004-08-05 Unigene Laboratories Inc. Improved oral delivery of peptides
WO2005012347A2 (en) 2003-08-05 2005-02-10 Novo Nordisk A/S Novel insulin derivatives
US20050282756A1 (en) 2004-06-18 2005-12-22 Mehta Nozer M Oral delivery of peptide pharmaceutical compositions
WO2008034881A1 (en) 2006-09-22 2008-03-27 Novo Nordisk A/S Protease resistant insulin analogues
GB2445013A (en) * 2006-11-29 2008-06-25 Malvern Cosmeceutics Ltd Novel compositions for the solubilisation of hydrophobic substances
WO2008145728A1 (en) 2007-06-01 2008-12-04 Novo Nordisk A/S Spontaneously dispersible preconcentrates including a peptide drug in a solid or semisolid carrier
WO2009115469A1 (en) 2008-03-18 2009-09-24 Novo Nordisk A/S Protease stabilized, acylated insulin analogues
WO2010060667A1 (en) * 2008-11-28 2010-06-03 Novo Nordisk A/S Pharmaceutical compositions suitable for oral administration of derivatized insulin peptides
WO2011068019A1 (en) 2009-12-02 2011-06-09 独立行政法人科学技術振興機構 Retrograde transport viral vector system having envelope comprising fused glycoprotein

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
"Eudragit@ Application Guidelines, 11th edition,", September 2009, EVONIK INDUSTRIES
"Handbook of Pharmaceutical Excipients,4th Edition", 2003, PHARMACEUTICAL PRESS
AMINO ACIDS, vol. 25, 2008, pages 233 - 241
DAVIES JT: "A quantitative kinetic theory of emulsion type, I. Physical chemistry of the emulsifying agent", GAS/LIQUID AND LIQUID/LIQUID INTERFACE. PROCEEDINGS OF THE INTERNATIONAL CONGRESS OF SURFACE ACTIVITY, 1957, pages 426 - 438, XP000869825
GREENE; WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
GRIFFIN WC: "Classification of Surface-Active Agents by'HLB", JOURNAL OF THE SOCIETY OF COSMETIC CHEMISTS, vol. 1, 1949, pages 311

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014060472A1 (en) * 2012-10-17 2014-04-24 Novo Nordisk A/S Fatty acid acylated amino acids for oral peptide delivery
WO2014060512A1 (en) * 2012-10-17 2014-04-24 Novo Nordisk Health Care Ag Fatty acid acylated amino acids for growth hormone delivery
WO2015162195A1 (en) * 2014-04-23 2015-10-29 Novo Nordisk A/S Fatty acid acylated amino acids for oral peptide delivery
EP4180060A1 (en) 2021-11-15 2023-05-17 Adocia Solid compositions comprising a peptide or a protein and an acylated amino acid
WO2023084118A1 (en) 2021-11-15 2023-05-19 Adocia Solid compositions comprising a peptide or a protein and an acylated amino acid
EP4299057A1 (en) 2022-06-30 2024-01-03 Adocia Solid compositions comprising a peptide or a protein and an acylated amino acid
EP4299071A1 (en) 2022-07-01 2024-01-03 Adocia Compositions comprising a peptide or a protein and an acylated amino acid
WO2024003400A1 (en) 2022-07-01 2024-01-04 Adocia Compositions comprising a peptide or a protein and an acylated amino acid

Also Published As

Publication number Publication date
US20140056953A1 (en) 2014-02-27
JP2014512366A (en) 2014-05-22
CN103458873A (en) 2013-12-18
EP2696847A1 (en) 2014-02-19
CN103458873B (en) 2016-04-13
JP6030630B2 (en) 2016-11-24

Similar Documents

Publication Publication Date Title
WO2012140155A1 (en) Fatty acid acylated amino acids for oral peptide delivery
JP5908847B2 (en) Pharmaceutical composition for oral administration of insulin peptides
Zupančič et al. Lipophilic peptide character–What oral barriers fear the most
US20150273069A1 (en) Fatty acid acylated amino acids for oral peptide delivery
AU2005328680B2 (en) Pharmaceutical compositions and methods for peptide treatment
EP2514406A1 (en) Spontaneously dispersible preconcentrates including a peptide drug in a solid or semisolid carrier
JP4785960B2 (en) Pharmaceutical composition
US20110293714A1 (en) Pharmaceutical compositions suitable for oral administration of derivatized insulin peptides
WO2014060512A1 (en) Fatty acid acylated amino acids for growth hormone delivery
CN101027318A (en) Insulin-oligomer conjugates, formulations and uses thereof
US20170258711A1 (en) Fatty Acid Acylated D-Amino Acids for Oral Peptide Delivery
WO2015162195A1 (en) Fatty acid acylated amino acids for oral peptide delivery
WO2022049310A1 (en) Improved pharmaceutical formulations of glp-1 receptor agonists
KR102216578B1 (en) Oral pharmaceutical composition comprising teriparatide and method for preparing the same
EP4181881A1 (en) Oral pharmaceutical compositions comprising lipid conjugates

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12713173

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2012713173

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2014504327

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14111274

Country of ref document: US