WO2015104310A1 - Stabilized pharmaceutical formulations of insulin aspart - Google Patents

Stabilized pharmaceutical formulations of insulin aspart Download PDF

Info

Publication number
WO2015104310A1
WO2015104310A1 PCT/EP2015/050215 EP2015050215W WO2015104310A1 WO 2015104310 A1 WO2015104310 A1 WO 2015104310A1 EP 2015050215 W EP2015050215 W EP 2015050215W WO 2015104310 A1 WO2015104310 A1 WO 2015104310A1
Authority
WO
WIPO (PCT)
Prior art keywords
insulin
pharmaceutical formulation
kit
pharmaceutical
inhibitors
Prior art date
Application number
PCT/EP2015/050215
Other languages
French (fr)
Inventor
Oliver BLEY
Petra Loos
Bernd Bidlingmaier
Walter Kamm
Harald Berchtold
Original Assignee
Sanofi
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 Sanofi filed Critical Sanofi
Priority to BR112016015851A priority Critical patent/BR112016015851A2/en
Priority to RU2016132340A priority patent/RU2016132340A/en
Priority to AU2015205620A priority patent/AU2015205620A1/en
Priority to SG11201604706TA priority patent/SG11201604706TA/en
Priority to JP2016544426A priority patent/JP6735674B2/en
Priority to CN201580003940.9A priority patent/CN105899190B/en
Priority to EP15700071.2A priority patent/EP3091964A1/en
Priority to CN202210571205.XA priority patent/CN114939156A/en
Priority to KR1020167021373A priority patent/KR20160101195A/en
Priority to CA2932873A priority patent/CA2932873A1/en
Priority to MX2016008977A priority patent/MX2016008977A/en
Publication of WO2015104310A1 publication Critical patent/WO2015104310A1/en

Links

Classifications

    • 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
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/30Zinc; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • 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
    • A61K38/26Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to a pharmaceutical formulation of insulin aspart, a process for preparing the pharmaceutical formulation of insulin aspart, and to a related kit. It also relates to the pharmaceutical formulation of insulin aspart and to the related kit for use in the treatment of diabetes mellitus, hyperglycemia, and/or for use in lowering blood glucose levels. The present invention also relates to the use of a medical device for administering the pharmaceutical formulation of insulin aspart to an animal and/or human.
  • Diabetes mellitus is a metabolic disorder in which the ability to utilize glucose is more or less completely lost.
  • insulin has been used in the treatment of diabetes mellitus.
  • insulin formulations e.g. insulin zinc (Zn (II) ) suspension, formulations containing protamine, etc.
  • the active pharmaceutical ingredient insulin itself has been modified by developing fast acting insulin analogues (e.g. insulin aspart, insulin lispro, insulin glulisine) and long acting insulin analogues and derivatives (e.g. insulin detemir, insulin degludec, insulin glargin).
  • fast acting insulin analogues e.g. insulin aspart, insulin lispro, insulin glulisine
  • long acting insulin analogues and derivatives e.g. insulin detemir, insulin degludec, insulin glargin.
  • Fast acting insulin preparations are usually solutions of insulin, while long acting insulin preparations can be suspensions containing insulin in crystalline and/or amorphous form precipitated by the addition of zinc (Zn(ll) ) salts alone or by addition of protamine or by a combination of both.
  • Insulin formulations are often administered by using pen injection devices or insulin pumps in which an insulin formulation is stored (in cartridges) until the entire cartridge is empty. Insulin formulations may also be stored in vials, requiring a stable formulation with respect to chemical and physical stability across the shelf life of the formulation.
  • the chemical and/or physical stability of insulin, insulin analogues and/or insulin derivatives strongly depends on the pharmaceutical formulation, e.g. the solvent, the pH value and the excipients.
  • Brange et al. (Acta Pharm. Nord. 4(3), pp. 149-158, 1992) disclose several aspects in connection with the chemical stability of insulin.
  • WO 2004/080480 discloses pharmaceutical preparations comprising acid-stabilized insulin.
  • GB 835,638 discloses insulin crystal suspensions having a protracted effect.
  • WO 98/56406 discloses stable insulin formulations.
  • US 6,489,292 discloses stable aqueous insulin preparations without phenol and cresol.
  • US 6,21 1 ,144 discloses stable concentrated insulin preparations for pulmonary delivery. Bhatt et al.
  • the German product specification of the medicinal product Berlinsulin ® H Normal 3ml_ Pen discloses a formulation containing human insulin, metacresol, glycerol, water and optionally hydrochloric acid and sodium hydroxide for pH adjustment.
  • the German product specification of the medicinal product Actrapid ® discloses a formulation containing human insulin, zinc chloride, glycerol, metacresol, water and optionally sodium hydroxide and hydrochloric acid for pH adjustment.
  • the FDA label of the medicinal product Lantus ® discloses a formulation containing insulin glargine, zinc, m-cresol, glycerol 85 %, polysorbate 20 and water for injection, wherein the pH is adjusted to approximately 4 by addition of aqueous solutions of hydrochloric acid and/or sodium hydroxide.
  • the FDA label of the medicinal product Humalog ® discloses a formulation containing insulin lispro, glycerin, dibasic sodium phosphate, metacresol, zinc oxide, phenol and water for injection, wherein the pH is adjusted to 7.0-7.8 by addition of aqueous solutions of hydrochlorid acid and/or sodium hydroxide.
  • the solubility of insulin, insulin analogues and/or insulin derivatives in aqueous media depends on the pH value. For example, the lowest solubility is shown close to the isoelectric point which for human insulin is around pH 5.3 and 5.4. Very good solubility can be observed at pH values below 4 and above 7. However, insulin suffers from degradation at strong acidic conditions and strong alkaline conditions. Therefore, most of the medicinal products containing insulin, insulin analogues and/or insulin derivatives have a pH value in the range of 7.2 to 7.4 and mostly buffering agents are used to achieve and maintain the pH within this range. It has now surprisingly been found that an alternative aqueous pharmaceutical formulation of insulin aspart having less than 0.17 mg/ml_ sodium chloride shows an excellent chemical and physical stability which qualifies this aqueous pharmaceutical formulation as a medicinal product having a defined shelf life.
  • One embodiment of the present invention relates to a pharmaceutical formulation comprising (a), insulin aspart; and
  • the pharmaceutical formulation contains no or less than 0.17 mg/ml_ sodium chloride.
  • the pharmaceutical formulation according to the present invention consists of (a), insulin aspart; and (b). Zn(ll); and (c). sodium chloride; and (d). optionally protamine; and (e). metacresol; (f). phenol; and (g). disodium phosphate (Na 2 HP0 4 ); (h). glycerol; and (i). sodium hydroxide and/or hydrochloric acid for pH adjustment to a pH value in the range of from 6.0 to 9.0; and (j)- water.
  • the pharmaceutical formulation according to the present invention is an aqueous pharmaceutical formulation.
  • the pharmaceutical formulation according to the present invention comprises at least one analogue and/or derivative of insulin which has or have an isoelectric point (lEP) from 4.0 to 6.0, from 4.5 to 6.0, from 4.5 to 5.5, from 5.0 to 5.5, from 5.0 to 5.2 or 5.1.
  • lEP isoelectric point
  • the pharmaceutical formulation according to the present invention has a pH value in the range from 6.0 to 9.0, from 6.5 to 8.5, from 7.0 to 8.0, from 7.0 to 7.8, from 7.1 to 7.6, or 7.2, 7.3, 7.4 or 7.5, or 7.4.
  • the pharmaceutical formulation according to the present invention comprises insulin aspart which is present in a concentration from 10 U/mL to 1000 U/mL, from 10 U/mL to 600 U/mL, from 10 U/mL to 300 U/mL, from 50 U/mL to 300 U/mL or 100 U/mL.
  • the pharmaceutical formulation according to the present invention comprises insulin aspart which is present in a concentration from 60 to 6000 nmol/mL, from 60 nmol/mL to 3600 nmol/mL, from 60 nmol/mL to 1800 nmol/mL, from 300 nmol/mL to 1800 nmol/mL or 600 nmol/mL.
  • the pharmaceutical formulation according to the present invention comprises Zn(ll) which is present in a concentration from 0.0100 mg/mL to 0.0600 mg/mL, from 0.0150 mg/mL to 0.0500 mg/mL, from 0.0150 mg/mL to 0.0300 mg/mL, from 0.0150 mg/mL to 0.0200 mg/mL, from 0.0190 mg/mL to 0.0200 mg/mL, or 0.0196 mg/mL.
  • the pharmaceutical formulation according to the present invention comprises zinc chloride (ZnCI 2 ) or zinc oxide (ZnO) or zinc acetate (anhydrous: C 4 H 6 0 4 Zn or dehydrate C 4 H 6 0 4 Zn x 2H 2 0).
  • the pharmaceutical formulation according to the present invention comprises Zn(ll) which is present in a concentration from 0.0100 mg/100 U to 0.0600 mg/100 U, from 0.0150 mg/100 U to 0.0500 mg/100 U, from 0.0150 mg/100 U to 0.0300 mg/100 U, from 0.0150 mg/100 U to 0.0200 mg/100 U, from 0.0190 mg/100 U to 0.0200 mg/100 U, or 0.0196 mg/100 U.
  • the pharmaceutical formulation according to the present invention comprises sodium chloride which is present in a concentration of less than 0.17 mg/mL, from 0 mg/mL to 0.17 mg/mL, from 0 mg/mL to 0.10 mg/mL, from 0 mg/mL to 0.05 mg/mL, or from 0 mg/mL to 0.01 mg/mL.
  • the pharmaceutical formulation according to the present invention does not contain any sodium chloride.
  • the pharmaceutical formulation according to the present invention is essentially free of sodium chloride.
  • the pharmaceutical formulation according to the present invention further contains a buffering agent.
  • the pharmaceutical formulation according to the present invention comprises protamine or protamine sulfate which is present in a concentration from 0.10, 0.15, 0.20, 0.25, 0.30, 0.32, 0.35, 0.40, 0.45 or 0.5 mg/mL.
  • the pharmaceutical formulation according to the present invention comprises a stabilizing agent, which is in one embodiment a surfactant, a polyoxyethylene derivative of sorbitan monolaurate (e.g. polysorbate 20), a polyethoxylethylene derivate of oleic acid (e.g.
  • polysorbate 80 poloxamer (which is a polyoxyethylene-polyoxypropylene copolymer), or polysorbate 20 or polysorbate 80 or mixtures thereof.
  • the stabilizing agent in one embodiment the surfactant, the polyoxyethylene derivative of sorbitan monolaurate (e.g. polysorbate 20), the polyethoxylethylene derivate of oleic acid (e.g.
  • polysorbate 80 poloxamer (which is a polyoxyethylene-polyoxypropylene copolymer), or polysorbate 20 or polysorbate 80 or mixtures thereof are/is present in a concentration from 0.01 to 0.05 mg/mLor in a concentration of 0.010 mg/mL, 0.015 mg/mL, 0.020 mg/mL, 0.025 mg/mL, 0.03 mg/mL, or 0.02 mg/mL.
  • the pharmaceutical formulation according to the present invention comprises insulin aspart and one or more further analogues and/or derivatives of insulin, including the following combinations
  • Insulin aspart and one or more analogues of insulin and in addition one or more derivative of insulin are provided.
  • the one or more analogue of insulin may be a fast acting insulin or a long acting insulin.
  • the one or more derivative of insulin may be a fast acting insulin or a long acting insulin.
  • the fast acting insulin is selected from the group comprising insulin aspart, insulin lispro and/or insulin glulisine.
  • the long acting insulin is selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec.
  • pharmaceutical formulation according to the present invention comprises insulin aspart and a long acting insulin (either an analogue or a derivative of insulin), wherein the long acting insulin is in one embodiment selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec.
  • the pharmaceutical formulation according to the present invention comprises one or more further active pharmaceutical ingredients.
  • the further active pharmaceutical ingredient is an antidiabetic agent.
  • the pharmaceutical formulation according to the present invention comprises one or more antidiabetic agents as further active pharmaceutical ingredients selected from the group comprising: GLP-1 receptor agonists, dual GLP-1 receptor/glucagon receptor agonists, human FGF-21 , FGF-21 analogues, FGF-21 derivatives, insulins, human insulin, analogues of insulin, and derivatives of insulin.
  • the pharmaceutical formulation according to the present invention comprises one or more further active pharmaceutical ingredients selected from the group comprising: insulin and insulin derivatives, GLP-1 , GLP-1 analogues and GLP-1 receptor agonists, polymer bound GLP-1 and GLP-1 analogues, dual GLP1/GIP agonists, dual GLP1/Glucagon receptor agonists, PYY3-36 or analogues thereof, pancreatic polypeptide or analogues thereof, glucagon receptor agonists or antagonists, GIP receptor agonists or antagonists, ghrelin antagonists or inverse agonists, Xenin and analogues thereof, DDP-IV inhibitors, SGLT2 inhibitors, dual SGLT2 / SGLT1 inhibitors, biguanides thiazolidinediones, dual PPAR agonists, sulfonylureas, meglitinides, alpha-glucosidase inhibitors, amylin and amylin analogue
  • pramlintide/metreleptin QNEXA (Phentermine+ topiramate), lipase inhibitors, angiogenesis inhibitors, H3 antagonists, AgRP inhibitors, triple monoamine uptake inhibitors (norepinephrine and acetylcholine), MetAP2 inhibitors, nasal formulation of the calcium channel blocker diltiazem, antisense oligonucleotides against production of fibroblast growth factor receptor 4, prohibitin targeting peptide-1 , drugs for influencing high blood pressure, chronic heart failure or atherosclerosis, such as angiotensin II receptor antagonists, ACE inhibitors, ECE inhibitors, diuretics, beta-blockers, calcium antagonists, centrally acting hypertensives, antagonists of the alpha-2-adrenergic receptor, inhibitors of neutral endopeptidase, thrombocyte aggregation inhibitors.
  • QNEXA Phhentermine+ topiramate
  • lipase inhibitors angiogenesis inhibitors
  • the pharmaceutical formulation according to the present invention consists of (a). 3.5 mg/mL insulin aspart; and (b). 1.72 mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). 0.0196 mg/mL Zn(ll); and (e).1.88 mg/mL Na 2 HP0 4 x 7 H 2 0 ; (f). 17.88 mg/mL glycerol; and (g). sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4 and (h). water.
  • the pharmaceutical formulation according to the present invention consists of (a). 3.5 mg/mL insulin aspart; and (b). 1.72 mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). 0.0196 mg/mL Zn(ll); and (e).1.88 mg/mL Na 2 HP0 4 x 7 H 2 0 ; (f). 17.88 mg/mL glycerol; (g). from 0.1 mg/mL to 0.5 mg/mL protamine sulfate; and (h). sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4 and (i). water.
  • the pharmaceutical formulation according to the present invention consists of (a). 3.5 mg/mL insulin aspart; and (b). 1.72 mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). 0.0196 mg/mL Zn(ll); and (e).1.88 mg/mL Na 2 HP0 4 x 7 H 2 0 ; (f). 17.88 mg/mL glycerol; (g). 0.1 or 0.15 or 0.2 or 0.25 or 0.3 or 0.32 or 0.35 or 0.4 or 0.45 or 0.5 mg/mL protamine sulfate; (h).
  • the present invention also provides to a pharmaceutical formulation according to the present invention for use in the treatment of diabetes mellitus, hyperglycemia and/or for use in lowering blood glucose levels.
  • the present invention also provides to a process for preparing the pharmaceutical formulation according to the present invention, wherein the components are mixed together in the form of a solution or suspension, the desired pH is adjusted and the mixture is made up to the final volume with water.
  • the present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention and of a medical device.
  • the medical device is selected from the group comprising: syringe, insulin injection system, insulin infusion system, insulin pump, insulin pen injection device.
  • the present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device.
  • the medical device is selected from the group comprising: syringe, insulin injection system, insulin infusion system, insulin pump, insulin pen injection device.
  • the present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the further active
  • the present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the further active
  • pharmaceutical ingredient is an antidiabetic agent selected from the group comprising: GLP-1 receptor agonists, dual GLP-1 receptor/glucagon receptor agonists, human FGF-21 , FGF-21 analogues, FGF-21 derivatives, insulins, human insulin, analogues of insulin, and derivatives of insulin.
  • the pharmaceutical formulation according to the present invention comprises one or more further active pharmaceutical ingredients selected from the group comprising: insulin and insulin derivatives, GLP-1 , GLP-1 analogues and GLP-1 receptor agonists, polymer bound GLP-1 and GLP-1 analogues, dual GLP1/GIP agonists, dual GLP1/Glucagon receptor agonists, PYY3-36 or analogues thereof, pancreatic polypeptide or analogues thereof, glucagon receptor agonists or antagonists, GIP receptor agonists or antagonists, ghrelin antagonists or inverse agonists, Xenin and analogues thereof, DDP-IV inhibitors, SGLT2 inhibitors, dual SGLT2 / SGLT1 inhibitors, biguanides thiazolidinediones, dual PPAR agonists, sulfonylureas, meglitinides, alpha-glucosidase inhibitors, amylin and amyl
  • the present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the kit comprises more than one analogue and/or derivative of insulin, wherein one analogue and/or derivative of insulin is a fast acting insulin and one analogue and/or derivative of insulin is a long acting insulin.
  • the fast acting insulin is selected from the group comprising insulin aspart, insulin lispro and/or insulin glulisine and wherein the long acting insulin is selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec.
  • the present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device for use in the treatment of diabetes mellitus, hyperglycemia and/or for use in lowering blood glucose levels.
  • the present invention also relates to a kit comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the further active pharmaceutical ingredient is an analogue or derivative of insulin selected from the group of fast acting insulin and long acting insulin.
  • the present invention also relates to a kit comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the further active pharmaceutical ingredient is an analogue or derivative of insulin selected from the group of fast acting insulin and long acting insulin and wherein the fast acting insulin is selected from the group comprising insulin aspart, insulin lispro and/or insulin glulisine and wherein the long acting insulin is selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec.
  • the further active pharmaceutical ingredient is an analogue or derivative of insulin selected from the group of fast acting insulin and long acting insulin and wherein the fast acting insulin is selected from the group comprising insulin aspart, insulin lispro and/or insulin glulisine and wherein the long acting insulin is selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec.
  • the present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the pharmaceutical formulation according to the present invention and the further active pharmaceutical ingredient, in one embodiment an antidiabetic agent, are administered continuously, separately, sequentially and/or stepwise.
  • the present invention also relates to the use of a medical device for administering the
  • the medical device is selected from the group comprising: syringe, insulin injection system, insulin infusion system, insulin pump, insulin pen injection device
  • active pharmaceutical ingredient includes any pharmaceutically active chemical or biological compound and any pharmaceutically acceptable salt thereof and any mixture thereof, that provides some pharmacologic effect and is used for treating or preventing a condition.
  • exemplary pharmaceutically acceptable salts include hydrochloric, sulfuric, nitric, phosphoric, hydrobromic, maleric, malic, ascorbic, citric, tartaric, pamoic, lauric, stearic, palmitic, oleic, myristic, lauryl sulfuric, naphthalinesulfonic, linoleic, linolenic acid, and the like.
  • active pharmaceutical ingredient “drug”, “active agent”, “active ingredient”, “active substance” and “drug” are meant to be synonyms, i.e., have identical meaning.
  • the active pharmaceutical ingredient is an antidiabetic agent.
  • antidiabetic agents are found in the Rote Liste 2012, chapter 12. Examples of antidiabetic agents include but are not Imitied to (a) insulin, insulin analogues and insulin derivatives, (b) glucagon-like- peptide 1 (GLP-1 ) and its analogues and receptor agonists, (c) dual GLP-1/GIP agonists, and (d) dual GLP-1 /glucagon receptor agonists, as described in detail next.
  • insulin, insulin analogues, and insulin derivatives include but are not limited to insulin glargine (Lantus ® ), insulin glulisine (Apidra ® ), insulin detemir (Levemir ® ), insulin lispro (Humalog ® / Liprolog ® ), insulin degludec (Tresiba ® ), insulin aspart (NovoLog ® / NovoRapid ® ), basal insulin and analogues (e.g.
  • LY2605541 , LY2963016 PEGylated insulin lispro, Humulin ® , Linjeta ® , SuliXen ® , NN1045, insulin plus Symlin ® , fast-acting and short-acting insulins (e.g. Linjeta ® , PH20 insulin, NN1218, HinsBet ® ), oral, inhalable, transdermal and sublingual insulins (e.g.
  • Exubera ® Nasulin ® , Afrezza ® , insulin tregopil, TPM -02/lnsulin, Capsulin ® , Oral-lyn ® , Cobalamin ® oral insulin, ORMD- 0801 , NN1953, VIAtab ® ). Additionally included are also those insulin derivatives which are bonded to albumin or another protein by a bifunctional linker.
  • GLP-1 Glucagon-like-peptide 1
  • GLP-1 analogues GLP-1 receptor agonists
  • GLP-1 , GLP-1 analogues and GLP-1 receptor agonists include but are not limited to lixisenatide (Lyxumia ® ), exenatide / exendin-4 (Byetta ® / Bydureon ® / ITCA 650, liraglutide / Victoza ® ), semaglutide, taspoglutide, albiglutide, dulaglutide, rExendin-4, CJC-1 134-PC, PB-1023, TTP-054, HM-1 1260C, CM-3, GLP-1 Eligen, ORMD-0901 , NN9924, Nodexen, Viador-GLP-1 , CVX- 096, ZYOG-1 , ZYD-1 , MAR-701 , ZP-2929, ZP-3022, CAM-2036, DA-15864, ARI-2651 , ARI-2255, exenatide-XTEN and glucagon-XTEN, AM
  • dual GLP-1/GIP agonists include but are not limited to MAR701 , MAR-709, BHM081 / BHM089 / BHM098).
  • Dual GLP-1 /glucagon receptor agonists include but are not limited to MAR701 , MAR-709, BHM081 / BHM089 / BHM098).
  • Examples of dual GLP-1/glucagon receptor agonists include but are not limited to OAP-189 (PF- 05212389, TKS-1225), TT-401/402, ZP2929, LAPS-HMOXM25, MOD-6030).
  • compositions of the invention include but are not limited to the following:
  • gastrointestinal peptides such as peptide YY 3-36 (PYY3-36) or analogues thereof and pancreatic polypeptide (PP) or analogues thereof.
  • Glucagon receptor agonists or antagonists GIP receptor agonists or antagonists, ghrelin antagonists or inverse agonists and xenin and analogues thereof.
  • Dipeptidyl peptidase-IV (DPP-4) inhibitors for example: alogliptin / Nesina ® , linagliptin / BI-1356 / Ondero ® / Trajenta ® / Tradjenta ® / Trayenta ® , saxagliptin / Onglyza ® , sitagliptin / Januvia ® / Xelevia ® / Tesavel ® , sitagliptin + metformin / Janumet ® / Velmetia ® , aildagliptin, anagliptin, aemigliptin, tenegliptin, melogliptin, trelagliptin, DA-1229, MK-3102, KM-223, KRP-104 and Ari-2243.
  • Sodium-dependent glucose transporter 2 (SGLT2) inhibitors for example: canagliflozin, dapagliflozin, remogliflozin, sergliflozin, empagliflozin, ipragliflozin, tofogliflozin (RO-4998452), luseogliflozin, LX-4211 , ertugliflozin (PF-04971729), EGT-0001442 and DSP-3235.
  • Biguanides e.g. metformin, buformin, phenformin
  • thiazolidinediones e.g. pioglitazone, rivoglitazone, rosiglitazone, troglitazone
  • dual PPAR agonists e.g. aleglitazar, muraglitazar, tesaglitazar
  • sulfonylureas e.g. tolbutamide, glibenclamide, glimepiride / Amaryl ® , glipizide
  • meglitinides e.g. nateglinide, repaglinide, mitiglinide
  • alpha-glucosidase inhibitors e.g. acarbose, miglitol, voglibose
  • amylin and amylin analogues e.g. pramlintide / Symlin ®
  • G-protein coupled receptor 119 GPR1 19 agonists (e.g. GSK-1292263, PSN-821 , MBX-2982, APD-597, ARRY-981 ).
  • GPR40 agonists e.g. TAK-875, TUG-424, P-1736, JTT-851 , GW9508.
  • GPR120 agonists and GPR142 agonists.
  • G-protein-coupled bile acid receptor 1 G-protein-coupled bile acid receptor 1 agonists (e.g. INT-777, XL-475, SB756050).
  • Bromocriptine / Cycloset ® inhibitors of 1 1 -beta-hydroxysteroid dehydrogenase (1 1 -beta-HSD) (e.g. LY2523199, BMS770767, RG-4929, BMS816336, AZD-8329, HSD-016, BI-135585), activators of glucokinase (e.g. PF-04991532, TTP-399, GK1 -399, ARRY-403 (AMG-151 ), TAK-329, ZYGK1 ), inhibitors of diacylglycerol O-acyltransferase (DGAT) (e.g.
  • pradigastat (LCQ-908), LCQ-908), inhibitors of protein tyrosinephosphatase 1 (e.g. trodusquemine), inhibitors of glucose-6- phosphatase, inhibitors of fructose-1 ,6-bisphosphatase, inhibitors of glycogen phosphorylase, inhibitors of phosphoenol pyruvate carboxykinase, inhibitors of glycogen synthase kinase, inhibitors of pyruvate dehydrogenase kinase, alpha2 adrenergic receptor antagonists, C-C chemokine receptor type 2 (CCR-2) antagonists, modulators of glucose transporter-4 and somatostatin receptor 3 agonists (e.g. MK ⁇ 1256).
  • protein tyrosinephosphatase 1 e.g. trodusquemine
  • inhibitors of glucose-6- phosphatase inhibitors of fructose-1 ,6-bisphosphatase
  • One or more lipid lowering agents are also suitable as active pharmaceutical ingredients, such as for example: 3-hydroxy-3-methylglutaryl-coenzym-A-reductase (HMG-CoA-reductase) inhibitors (e.g. simvastatin, atorvastatin, rosuvastatin), fibrates (e.g. bezafibrate, fenofibrate), nicotinic acid and derivatives thereof (e.g. niacin, including slow release formulations of niacin), nicotinic acid receptor 1 agonists (e.g.
  • HMG-CoA-reductase) inhibitors e.g. simvastatin, atorvastatin, rosuvastatin
  • fibrates e.g. bezafibrate, fenofibrate
  • nicotinic acid and derivatives thereof e.g. niacin, including slow release formulations of niacin
  • PPAR- peroxisome proliferator-activated receptors
  • ACAT acetyl- CoA-acetyltransferase
  • bile acid-binding substances e.g. cholestyramine, colesevelam
  • ileal bile acid transport inhibitors IBAT
  • MTP microsomal triglyceride transfer protein
  • AEGR-733 e.g. lomitapide (AEGR-733)
  • SLx-4090 granotapide
  • PCSK9 subtilisin/kexin type 9
  • AMG-145 subtilisin/kexin type 9
  • PF-04950615 PF-04950615
  • MPSK3169A LY3015014
  • ALD-306 ALN-PCS
  • BMS-962476 SPC5001
  • ISIS-394814 1 B20
  • LGT-210 1 D05
  • BMS-PCSK9Rx-2 SX-PCK9
  • RG7652 LDL receptor up-regulators
  • liver selective thyroid hormone receptor beta agonists e.g.
  • eprotirome (KB-21 15), MB0781 1 , sobetirome (QRX-431 ), VIA-3196, ZYT1 ), HDL-raising compounds such as: CETP inhibitors (e.g. torcetrapib, anacetrapib (MK0859), dalcetrapib, evacetrapib, JTT-302, DRL-17822, TA-8995, R- 1658, LY-2484595) or ABC1 regulators, lipid metabolism modulators (e.g. BMS-823778, TAP-301 , DRL-21994, DRL-21995), phospholipase A2 (PLA2) inhibitors (e.g.
  • CETP inhibitors e.g. torcetrapib, anacetrapib (MK0859), dalcetrapib, evacetrapib, JTT-302, DRL-17822, TA-8995, R- 1658, LY-2484595
  • darapladib / Tyrisa ® varespladib, rilapladib
  • ApoA-l enhancers e.g. RVX-208 , CER-001 , MDCO-216, CSL-1 12, VRX- HDL, VRX-1243, VIRxSYS
  • cholesterol synthesis inhibitors e.g. ETC-1002
  • lipid metabolism modulators e.g. BMS-823778, TAP-301 , DRL-21994, DRL-21995
  • omega-3 fatty acids and derivatives thereof e.g. icosapent ethyl (AMR101 ), Epanova ® , AKR-063, NKPL-66).
  • compositions include one or more active substances for the treatment of obesity, including but not limited to:
  • Sibutramine, tesofensine, orlistat, cannabinoid receptor 1 (CB1 ) antagonists e.g. TM-38837
  • MH-1 melanin-concentrating hormone
  • MH-1 melanin-concentrating hormone
  • MC4 receptor agonists and partial agonists e.g. AZD-2820, RM-493
  • NPY5 neuropeptide Y5
  • NPY2 antagonists e.g. velneperit, S-234462
  • NPY4 agonists e.g.
  • PP-1420 beta-3-adrenergic receptor agonists, leptin or leptin mimetics, agonists of the 5-hydroxytryptamine 2c (5HT2c) receptor (e.g. lorcaserin), or the combinations of bupropione/naltrexone (Contrave ® ),
  • bupropione/zonisamide (Empatic ® ), bupropione/phentermine or pramlintide/metreleptin, phentermine/topiramate (Qsymia ® ), lipase inhibitors (e.g. cetilistat / Cametor ® ), angiogenesis inhibitors (e.g. ALS-L1023), histamine H3 antagonists (e.g. HPP-404), AgRP (agouti related protein) inhibitors (e.g. TTP-435), triple monoamine uptake inhibitors (dopamine, norepinephrine and serotonin reuptake) (e.g.
  • tesofensine methionine aminopeptidase 2 (MetAP2) inhibitors (e.g. beloranib), nasal formulations of the calcium channel blocker diltiazem (e.g. CP-404) and antisense oligonucleotides against production of fibroblast growth factor receptor 4 (FGFR4) (e.g. ISIS- FGFR4Rx) or prohibitin targeting peptide-1 (e.g. Adipotide ® ).
  • FGFR4 fibroblast growth factor receptor 4
  • FGFR4 fibroblast growth factor receptor 4
  • prohibitin targeting peptide-1 e.g. Adipotide ®
  • compositions include but are not limited to:
  • Angiotensin II receptor antagonists e.g. telmisartan, candesartan, valsartan, losartan, eprosartan, irbesartan, olmesartan, tasosartan, azilsartan
  • angiotensin converting enzyme (ACE) inhibitors e.g. telmisartan, candesartan, valsartan, losartan, eprosartan, irbesartan, olmesartan, tasosartan, azilsartan
  • ACE angiotensin converting enzyme
  • EAE endothelin converting enzyme
  • diuretics e.g., beta-blockers, calcium antagonists, centrally acting hypertensives, antagonists of the alpha-2-adrenergic receptor, inhibitors of neutral endopeptidase, thrombocyte aggregation inhibitors and others or combinations thereof are suitable.
  • analogue of insulin refers to a polypeptide which has a molecular structure which formally can be derived from the structure of a naturally occurring insulin, for example that of human insulin, by deleting and/or exchanging at least one amino acid residue occurring in the naturally occurring insulin and/or adding at least one amino acid residue.
  • the added and/or exchanged amino acid residue can either be codable amino acid residues or other naturally occurring residues or purely synthetic amino acid residues.
  • analogues of insulin include, but are not limited to, the following: (i). ' Insulin aspart ' is created through recombinant DNA technology so that the amino acid B28 in human insulin (i.e. the amino acid no.
  • Insulin lispro ' is created through recombinant DNA technology so that the penultimate lysine and proline residues on the C-terminal end of the B-chain of human insulin are reversed (human insulin: Pro B28 Lys B29 ; insulin lispro: Lys B28 Pro B29 ); (iii). ' Insulin glulisine ' differs from human insulin in that the amino acid asparagine at position B3 is replaced by lysine and the lysine in position B29 is replaced by glutamic acid; (iv).
  • “Insulin glargine” differs from human insulin in that the asparagine at position A21 is replaced by glycine and the B chain is extended at the carboxy terminal by two arginines.
  • aqueous refers to a solution in which the solvent is water and/or to a suspension in which the external phase is water and/or to an emulsion in which the dispersed or continuous phase is water.
  • buffering agent refers to a weak acid or base used to maintain the acidity (pH) of a solution, a suspension and/or an emulsion near a chosen value after the addition of another acid or base.
  • a buffering agent is to prevent a rapid change in the pH value when acids or bases are added to the solution.
  • a buffering agent is present in a mixture of a weak acid and its conjugate base or a in a mixture of a weak base and its conjugated acid.
  • buffering agents include, but are not limited to, the following: sodium bicarbonate; acetic acid or acetate salts (e.g.
  • boric acid or boric salts N-cyclohexyl-2-aminoethanesulfonic acid (CHES) or salts thereof; 3-[[1 ,3-dihydroxy-2-(hydroxymethyl)propan-2-yl]amino]propane-1-sulfonic acid (TAPS) or salts thereof; 2-(N-morpholino)ethanesulfonic acid (MES) and salts therof; piperazine- ⁇ , ⁇ -bis(2- ethanesulfonic acid (PIPES) and salts therof; N-(2-acetamido)-2-aminoethane-sulfonic acid (ACES) and salts therof; cholamine chloride; BES; 2-[[1 ,3-dihydroxy-2-(hydroxymethyl)-propan-2- yl]amino]ethanesulfonic acid (TES) and salts therof; 2-[4-(2-hydroxye
  • sodium citrate trisodium phosphate, disodium hydrogen phosphate, sodium dihydrogen phosphate, tripotassium phosphate, dipotassium phosphate, monopotassium phosphate and/or any other buffering agent containing phosphate.
  • buffering agent also comprises amino acids, peptides and proteins.
  • insulin analogues and/or insulin derivatives and/or protamine are peptides or derivatives of peptides (i.e. both contain amino acids having free basic or acidic functional groups), they may also have a certain buffering capacity, i.e. are also to be considered as buffering agent.
  • fast acting insulin refers to insulin analogues and/or insulin derivatives, wherein the insulin-mediated effect begins within 5 to 15 minutes and continues to be active for 3 to 4 hours.
  • fast acting insulins include, but are not limited to, the following: (i). insulin aspart; (ii). insulin lispro and (iii). insulin glulisine.
  • the terms ..derivative of insulin” and "insulin derivative” refers to a polypeptide which has a molecular structure which formally can be derived from the structure of a naturally occurring insulin, for example that of human insulin, in which one or more organic substituent (e.g. a fatty acid) is bound to one or more of the amino acids.
  • one or more amino acids occurring in the naturally occurring insulin may have been deleted and/or replaced by other amino acids, including non-codeable amino acids, or amino acids, including non-codeable, have been added to the naturally occurring insulin.
  • derivatives of insulin include, but are not limited to, the following: (i).
  • Insulin detemir ' which differs from human insulin in that the C-terminal threonine in position B30 is removed and a fatty acid residue (myristic acid) is attached to the epsilon-amino function of the lysine in position B29.
  • Insulin degludec ' which differs from human insulin in that the last amino acid is deleted from the B-chain and by the addition of a glutamyl link from Lys B29 to a hexadecandioic acid.
  • FGF-21 means "fibroblast growth factor 21 ".
  • FGF-21 compounds may be human FGF-21 , an analogue of FGF-21 (referred to "FGF-21 analogue") or a derivative of FGF-21 (referred to "FGF-21 derivative”).
  • FGF-21 derivative a derivative of FGF-21
  • formulation refers to a product comprising specified ingredients in predetermined amounts or proportions, as well as any product that results, directly or indirectly, from combining specified ingredients in specified amounts. In relation to pharmaceutical
  • formulations this term encompasses a product comprising one or more active ingredients, and an optional carrier comprising inert ingredients, as well as any product that results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • pharmaceutical formulations are prepared by uniformly bringing the active pharmaceutical ingredient (i.e. the analogue and/or derivative of insulin) into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the pharmaceutical formulation includes enough of the active pharmaceutical ingredient to produce the desired effect upon the progress or condition of diseases.
  • formulation may refer to a solution as well as to a suspension or to an emulsion.
  • formulation and “composition” are meant to be synonyms, i.e., have identical meaning.
  • the pharmaceutical compositions are made following conventional techniques of pharmaceutical technology involving mixing, filling and dissolving the ingredients, as appropriate, to give the desired oral, parenteral, rectal, transdermal, or topical products.
  • GLP-1 receptor agonist refers to compounds which have an agonistic activity at the glucagon-like peptide-1 receptor.
  • GLP-1 receptor agonists include, but are not limited to, the following: exenatide / exendin-4, liraglutide, lixisenatide, dulaglutide, albiglutide, semaglutide, taspoglutide, rExendin-4, CJC-1 134-PC, PB-1023, TTP-054, HM-1 1260C, CM-3, GLP-1 Eligen, ORMD-0901 , NN9924, Nodexen, Viador-GLP-1 , CVX-096, ZYOG-1 , ZYD-1 , MAR-701 , ZP-2929, ZP-3022, CAM-2036, DA-15864, ARI-2651 , ARI-2255, exenati de-XT EN and glucagon-XTEN, AMX-8089+
  • dual GLP-1 receptor/glucagon receptor agonist refers to compounds which have agonistic activity at both the GLP-1 receptor and the glucacon receptor.
  • dual GLP-1 receptor/glucagon receptor agonist include, but are not limited to, the following:
  • human insulin refers to the human hormone whose structure and properties are well-known. Human insulin has two polypeptide chains (chains A and B) that are connected by disulphide bridges between cysteine residues, namely the A-chain and the B-chain.
  • the A-chain is a 21 amino acid peptide and the B-chain is a 30 amino acid peptide, the two chains being connected by three disulphide bridges: one between the cysteins in position 6 and 1 1 of the A-chain; the second between the cysteine in position 7 of the A-chain and the cysteine in position 7 of the B-chain; and the third between the cysteine in position 20 of the A-chain and the cysteine in position 19 of the B-chain.
  • the term "isoelectric point" refers to the pH value at which a particular molecule carries no net electrical charge.
  • the isoelectric point can be determined by using isoelectric focusing, which is a technique for separating different molecules by differences in their isoelectric point and which is well known in the art. It can also be calculated (see e.g. Levene and Simms, ' Calculation of isoelectric point ' J. Biol. Chem., 1923, pp. 801 -813).
  • the term “kit” refers to a product (e.g. medicament, kit-of-parts) comprising one package or one or moreseparate packages of:
  • a pharmaceutical formulation containing an active pharmaceutical ingredient and at least one further active pharmaceutical ingredient and optionally a medical device may be present in said pharmaceutical formulation, i.e. the kit may comprise one or more packages, wherein each package comprises one pharmaceutical formulation which comprises two or more active pharmaceutical ingredients.
  • the further active pharmaceutical ingredient may also be present in a further pharmaceutical formulation, i.e. the kit may comprise separate packages of two or more pharmaceutical formulations, wherein each pharmaceutical formulation contain one active pharmaceutical ingredient.
  • a pharmaceutical formulation containing an active pharmaceutical ingredient and medical device (ii) .
  • a kit may comprise one package only or may comprise one or more separate packages
  • the kit may be a product (e.g. medicament) containing two or more vials each containing a defined pharmaceutical formulation, wherein each pharmaceutical formulation contains at least one active pharmaceutical ingredient.
  • the kit may comprise (i.) a vial containing a defined pharmaceutical formulation and (ii). further a tablet, capsule, powder or any other oral dosage form which contains at least one further active pharmaceutical ingredient.
  • the kit may further comprise a package leaflet with instructions for how to administer the pharmaceutical formulation and the at least one further active pharmaceutical ingredient.
  • the term “medical device” means an instrument, apparatus, implant, in vitro reagent or similar or related article that is used to diagnose, prevent, or treat a disease of other condition, and does not achieve its purpose through pharmacological action within or on the body.
  • a medical device may be a syringe, an insulin injection system, an insulin infusion system, an insulin pump or an insulin pen injection device.
  • a medical device may be mechanically or electromechanically driven.
  • the conjunction "or” is used in the inclusive sense of "and/or” and not the exclusive sense of "either/or”.
  • pH and "pH value” refer to the decimal logarithm of the reciprocal of the hydrogen ion activity in a solution.
  • the term “pharmaceutical” refers to the intended use in the medical diagnosis, cure, treatment and/or prevention of diseases.
  • pharmaceutically acceptable refers to physiologically well tolerated by a mammal or a human.
  • protamine refers to a mixture of strongly basic peptides. It was originally isolated from the sperm of salmon and other species of fish but is now produced primarily recombinant through biotechnology. It contains more than two-thirds of L-arginine. As protamine contains amino acids having free basic side chains, it has a certain buffering capacity and is therefore considered to be a buffering agent. Protamine may be used as protamine sulfate or protamine hydrochloride. Concentrations, amounts, solubility in different liquid systems, particle size, wavelength, pH values, weight mass, molecular weight, percent and other numerical date may be expressed or presented herein in a range format.
  • long acting insulin refers to insulin analogues and/or insulin derivatives, wherein the insulin-mediated effect begins within 0.5 to 2 hours and continues to be active, for about or more than 24 hours.
  • fast acting insulins include, but are not limited to, the following: (i). insulin glargin; (ii). insulin detemir and (iii). insulin degludec.
  • the term "stability" refers to the chemical and/or physical stability of active pharmaceutical ingredients, in particular of insulin analogues and/or derivatives.
  • the purpose of stability testing is to provide evidence on how the quality of an active pharmaceutical ingredient or dosage form varies with time under the influence of a variety of environmental factors such as temperature, humidity, and light, and to establish a shelf life for the active pharmaceutical ingredient or dosage form and recommended storage conditions.
  • Stability studies can include testing of those attributes of the active pharmaceutical ingredient that are susceptible to change during storage and are likely to influence quality, safety, and/or efficacy.
  • the testing can cover, as appropriate, the physical, chemical, biological, and microbiological attributes, preservative content (e.g., antioxidant, antimicrobial preservative), and functionality tests (e.g. for a dose delivery system).
  • Analytical procedures can be fully validated and stability indicating. In general, significant changes for an active pharmaceutical ingredient and/or dosage form with regard to stability are defined as:
  • the significant changes may also be evaluated against established acceptance criteria prior to starting the evaluation of the stability.
  • Acceptance criteria can be derived from the monographs (e.g. monographs for the European Pharmacopeia, of the United States Pharmacopeia, of the British Pharmacopeia, or others), and from the analytical batches of the active pharmaceutical ingredient and medicinal product used in the preclinical and clinical studies. Acceptable limits should be proposed and justified, taking into account the levels observed in material used in preclinical and clinical studies. Product characteristics may be visual appearance, purity, color and clarity for solutions/suspensions, visible particulates in solutions, and pH. As a non-limiting example, suitable acceptance criteria for insulin aspart formulations are shown below:
  • Assay insulin aspart units 90.0 insulin aspart units/mL to 1 10.0 insulin
  • the acceptance criteria shown above are based on monographed acceptance limits (e.g. British Pharmacopoeia, Volume III, 2012 or Pharmacopoeial Forum, Volume 36(6), Nov-Dec 2010) and/or are derived from extensive experience in the development of insulin formulations.
  • treatment refers to any treatment of a mammalian, for example human condition or disease, and includes: (1 ) inhibiting the disease or condition, i.e., arresting its development, (2) relieving the disease or condition, i.e., causing the condition to regress, or (3) stopping the symptoms of the disease.
  • the unit of measurement grillU" and/or Jnternational units refers to the blood glucose lowering activity of insulin and is defined (according to the World Health Organization, WHO) as follows: 1 U corresponds to the amount of highly purified insulin (as defined by the WHO) which is sufficient to lower the blood glucose level of a rabbit (having a body weight of 2 - 2.5 kg) to 50 mg / 100 ml. within 1 hour and to 40 mg / 100 ml. within 2 hours.
  • 1 U corresponds to approximately 35 ⁇ g (Lill, Pharmazie in 102, No. 1 , pp. 56-61 , 2001 ).
  • 100 U correspond to 3.5 mg (product information NovoRapid ® ).
  • For insulin lispro 100 U correspond to 3.5 mg (product information Humalog ® ).
  • For insulin glulisine 100 U correspond to 3.49 mg (product information Apidra ® cartridges).
  • For insulin determir 100 U correspond to 14.2 mg (product information Levemir ® ).
  • For insulin glargin 100 U correspond to 3.64 mg (product information Lantus ® ). Further embodiments of the present invention include the following:
  • the invention provides a pharmaceutical formulation comprising (a), insulin aspart; and (b). Zn(ll); and (c). optionally protamine; wherein the pharmaceutical formulation contains less than 0.17 mg/ml_ sodium chloride.
  • the pharmaceutical formulation of the invention is an aqueous pharmaceutical formulation.
  • the pharmaceutical formulation of the invention has a pH value in the range from 6.0 to 9.0.
  • the pharmaceutical formulation of the invention has a pH value in the range from 7.0 to 7.8.
  • the pharmaceutical formulation of the invention comprises insulin aspart which is present in a concentration from 10 U/mL to 1000 U/mL.
  • the pharmaceutical formulation of the invention comprises Zn(ll) which is present in a concentration from 0.0100 to 0.0600 mg / 100 U of insulin aspart.
  • the pharmaceutical formulation of the invention is essentially free of sodium chloride.
  • the pharmaceutical formulation of the invention comprises protamine which is present in a concentration from 0.1 to 0.5 mg/ml_.
  • the pharmaceutical formulation of the invention further contains a buffering agent.
  • the pharmaceutical formulation of the invention comprises one or more further active pharmaceutical ingredients.
  • the pharmaceutical formulation of the invention comprises a further active pharmaceutical ingredient which is an antidiabetic agent.
  • the pharmaceutical formulation of the invention comprises a further active pharmaceutical ingredient which is an antidiabetic agent selected from the group consisting of: (a), a GLP-1 receptor agonist; (b). a dual GLP-1 receptor/glucagon receptor agonist; (c). human FGF-21 ; (d). a FGF-21 analogue; (e). a FGF-21 derivative; (f). insulin; (g). human insulin;
  • the pharmaceutical formulation of the invention further comprises a fast acting analogue and/or derivative of insulin or a long acting analogue and/or derivative of insulin.
  • the pharmaceutical formulation of the invention comprises a fast acting analogue and/or derivative of insulin or a long acting analogue and/or derivative of insulin, wherein the fast acting insulin is one or more insulin selected from the group consisting of insulin lispro and insulin glulisine and wherein the long acting insulin is one or more insulin selected from the group consisting of insulin detemir, insulin glargin and insulin degludec.
  • the pharmaceutical formulation of the invention consists of: (a). 3.5 mg/ml_ insulin aspart; (b). 0.0196 mg/mL Zn(ll) ; (c). 1.88 mg/mL Na 2 HP0 4 x 7 H 2 0; (d). 1.72 mg/mL m-cresol; (e). 1.5 mg/mL phenol; (f). 17.88 mg/mL glycerol; (g). sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4; and (h). water.
  • the pharmaceutical formulation of the invention consists of: (a). 3.5 mg/mL insulin aspart; (b). 0.0196 mg/mL Zn(ll); (c). 1.88 mg/mL Na 2 HP0 4 x 7 H 2 0; (d). 1.72 mg/mL m-cresol; (e). 1.5 mg/mL phenol; (f). 17.88 mg/mL glycerol; (g). from 0.1 mg/mL to 0.5 mg/mL protamine sulfate; (h). sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4; and (i). water.
  • the invention provides a process for preparing the pharmaceutical formulation of the invention, wherein the components are mixed together in the form of a solution or suspension, the pH is adjusted to reach the desired pH, and water is added to reach the final volume.
  • the invention provides a kit comprising one or more separate packages of (a), the pharmaceutical formulation of the invention; and (b). a medical device.
  • the invention provides as kit comprising one or more separate packages of (a), the pharmaceutical formulation if the invention; and (b). at least one further active pharmaceutical ingredient; (c). and optionally a medical device.
  • the kit of the invention comprises a further active pharmaceutical ingredient which is an antidiabetic agent.
  • the kit of the invention comprises a further active pharmaceutical ingredient which is an antidiabetic agent selected from the group consisting of: (a), a GLP-1 receptor agonist;
  • the kit of the invention comprises a further active pharmaceutical ingredient which is an analogue or derivative of insulin selected from the group consisting of fast acting insulin and long acting insulin.
  • the kit of the invention comprises an analogue or derivative of insulin selected from the group consisting of fast acting insulin and long acting insulin, wherein the fast acting insulin is selected from the group consisting of insulin aspart, insulin lispro and insulin glulisine and wherein the long acting insulin is selected from the group consisting of insulin glargin, insulin detemir and insulin degludec.
  • the invention provides a pharmaceutical formulation or a kit for use in the treatment of diabetes mellitus.
  • the invention provides a pharmaceutical formulation or a kit for use in the treatment of hyperglycemia.
  • the invention provides a pharmaceutical formulation or a kit for use in lowering blood glucose level. In one aspect, the invention provides a method of treating diabetes mellitus in a subject in need thereof comprising administering the pharmaceutical formulation of the invention.
  • the invention provides a method of treating hyperglycemia in a subject in need thereof comprising administering the pharmaceutical formulation of the invention. In one aspect, the invention provides a method of lowering blood glucose levels in a subject in need thereof comprising administering the pharmaceutical formulation of the invention.
  • the invention provides a medical device for administering the pharmaceutical formulation of the invention to an animal and/or human.
  • Zinc oxide Solution (containing Zn(ll) ) was prepared by suspending 0.8539 g zinc oxide in 500 mL water for injection and dissolving by adding 1 N hydrochloric acid. The solution is filled up with water for injection to final volume of 1000 mL. (b) Solution A
  • Solution A was prepared as described in the following:
  • Solution was filled up to approximately 1800 g with water for injection.
  • pH value was changed to approximately 3.1 to 3.2 by adding hydrochloric acid 0.03 N or sodium hydroxide solution 0.02 N to dissolve the insulin aspart.
  • Solution was filled up to 600 g with water for injection.
  • pH was adjusted to 7.4 (range 7.2 to 7.6) using hydrochloric acid 0.03 N or sodium
  • hydroxide solution 0.02 N. 10.
  • Solution was filled up to 2010 g with water for injection (corresponds to 100 % of the Final Solution).
  • Final solution was a clear and uncoloured solution, showed a pH value of 7.4 (plus/minus 0.2; at 20-25°C).
  • the Final Solution was applied to sterile filtration using ' Sartopore Minisart high flow ' filter (filter material: polyethersulfone; pore size: 0.2 ⁇ ; supplier: Sartorius).
  • the Final Solution after sterile filtration was a clear and uncoloured solution and showed an osmolarity of 260 mOsmol/kg (plus/minus 30).
  • the Final Solution after sterile filtration was filled into appropriate vials (volume: 5 and 10 mL; 13 mm; clear glas; glas type 1 ).
  • the vials -containing the Final Solution after sterile filtration- were stored between +2°C and + 8°C and protected from light.
  • Tests are carried out using compendial analytical test methods, where applicable.
  • the quality control concept has been established taking into account the cGMP requirements as well as the current status of the ICH process.
  • the identity of the active ingredient is ensured by comparing the retention time of the drug formulation sample with the retention time of the reference standard using a reversed phase HPLC method.
  • the method is also used for the determination of assay of the active ingredient, for the determination of the related compounds and impurities, and for quantifying the preservatives m- cresol and phenol.
  • the test is carried out by reverse phase liquid chromatography (HPLC).
  • HPLC reverse phase liquid chromatography
  • the method is also used for the identification, the determination of assay of the active ingredient, for the determination of the related compounds and impurities, and for quantifying the preservatives m-cresol and phenol.
  • Autosampler Thermostated at ⁇ +8 °C.
  • Mobile phase A Sodium sulfate solved in water, 14 g/mL, adjusted with phosphoric acid and sodium hydroxide to a pH of 3.4.
  • Mobile phase B Water/acetonitrile (50:50 v/v). Gradient is shown in Table 3.
  • Impurities are calculated using the peak area percent method.
  • Test solution The formulation is used without any dilution or further treatment.
  • Related compounds and impurities HPLC
  • High molecular weight proteins HMWPs
  • the high molecular weight proteins are determined using high pressure size exclusion
  • HMWPs are calculated using the peak area percent method.
  • Test solution The formulation is used without any dilution or further treatment.
  • HPLC analytical procedure for the formulation for the determination of identification, assay, and related compounds and impurities was validated to demonstrate specificity, linearity, limit of detection and limit of quantification, accuracy, precision and range.
  • Stability studies for the formulation were initiated according to the stability protocol summary described in the following table.
  • the composition and manufacturing method of the stability batches are representative of the material.
  • the stability profile was assessed for storage under long term, accelerated, and stress testing conditions according to ICH guidelines.
  • Samples were packed and stored in glass vials with flanged cap with inserted disc and flip-off lid.
  • the stability data obtained using this packaging material are representative for the preliminary shelf life and storage direction for both packaging configurations (10 ml. glass vials and 3 ml. cartridges).
  • Tables 5 and 6 show the long term stability results, wherein batch no. "_0021 " is referring to a formulation according to the present invention.
  • the stability of the formulation as presently claimed shows an excellent chemical and physical stability which qualifies said aqueous pharmaceutical formulation as medicinal product having a defined shelf life.
  • Table 5 Stability data of batch _0021 stored for 1 month at long term storage condition +5°C, at accelerated condition +25°C, and at stress condition +40°C
  • insulin aspart 90.0 insulin aspart 107.7 insulin 108.0 insulin 91.4 insulin 102.9 insulin 96.0 insulin aspart units (HPLC) units/mL to 110.0 insulin aspart units/mL aspart units/mL aspart aspart units/mL units/mL aspart units/mL (3.77 mg/mL) (3.78 mg/mL) units/mL (3.60 mg/mL) (3.36 mg/mL)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Inorganic Chemistry (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Dermatology (AREA)
  • Obesity (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Hematology (AREA)
  • Emergency Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Infusion, Injection, And Reservoir Apparatuses (AREA)

Abstract

Stabilized pharmaceutical formulations of insulin aspart are disclosed. In particular the application is directed to a pharmaceutical formulation comprising (a) insulin aspart; and (b) Zn(ll); and (e) optionally protamine; wherein the pharmaceutical formulation contains less than 0.17 mg/mL sodium chloride.

Description

Stabilized pharmaceutical formulations of insulin aspart. INTRODUCTION
The present invention relates to a pharmaceutical formulation of insulin aspart, a process for preparing the pharmaceutical formulation of insulin aspart, and to a related kit. It also relates to the pharmaceutical formulation of insulin aspart and to the related kit for use in the treatment of diabetes mellitus, hyperglycemia, and/or for use in lowering blood glucose levels. The present invention also relates to the use of a medical device for administering the pharmaceutical formulation of insulin aspart to an animal and/or human.
BACKGROUND OF THE INVENTION
Diabetes mellitus is a metabolic disorder in which the ability to utilize glucose is more or less completely lost.
For decades, insulin has been used in the treatment of diabetes mellitus. Several insulin formulations have been developed, e.g. insulin zinc (Zn (II) ) suspension, formulations containing protamine, etc. Further, the active pharmaceutical ingredient insulin itself has been modified by developing fast acting insulin analogues (e.g. insulin aspart, insulin lispro, insulin glulisine) and long acting insulin analogues and derivatives (e.g. insulin detemir, insulin degludec, insulin glargin). Fast acting insulin preparations are usually solutions of insulin, while long acting insulin preparations can be suspensions containing insulin in crystalline and/or amorphous form precipitated by the addition of zinc (Zn(ll) ) salts alone or by addition of protamine or by a combination of both.
The chemical and physical stability of insulin formulations is very important. Insulin formulations are often administered by using pen injection devices or insulin pumps in which an insulin formulation is stored (in cartridges) until the entire cartridge is empty. Insulin formulations may also be stored in vials, requiring a stable formulation with respect to chemical and physical stability across the shelf life of the formulation.
The chemical and/or physical stability of insulin, insulin analogues and/or insulin derivatives strongly depends on the pharmaceutical formulation, e.g. the solvent, the pH value and the excipients. Brange et al. (Acta Pharm. Nord. 4(3), pp. 149-158, 1992) disclose several aspects in connection with the chemical stability of insulin. WO 2004/080480 discloses pharmaceutical preparations comprising acid-stabilized insulin. GB 835,638 discloses insulin crystal suspensions having a protracted effect. WO 98/56406 discloses stable insulin formulations. US 6,489,292 discloses stable aqueous insulin preparations without phenol and cresol. US 6,21 1 ,144 discloses stable concentrated insulin preparations for pulmonary delivery. Bhatt et al. (Pharmaceutical Research, Vol. 7, No. 6, pp. 593-599, 1990) disclose chemical pathways of peptide degradation. Patel et al. (Pharmaceutical Research, Vol. 7, No. 7, pp. 703-71 1 , 1990) disclose chemical pathways of peptide degradation. Tyler-Cross et al. (The Journal of Biological Chemistry, Vol. 266, No. 33, Issue of November 25, pp. 22549-22556, 1991 ) disclose effects of amino acid sequence, buffers, and ionic strength on the rate and mechanism of deamidation of asparagine residues in small peptides. GB 840,870 discloses improvements in or relating to insulin preparations. US
6,852,694 discloses stabilized insulin formulations. Galloway et al. (Diabetes - The Journal of the American Diabetes Association, Vol. 21 , No. Suppl.2, pp. 637-648, 1972) disclose new forms of insulin. Jackson et al. disclose several aspects with regard to neutral regular insulin (Diabetes - The Journal of the American Diabetes Association, Vol. 21 , No. 4, pp. 235-245, 1972). Lill (Pharmazie in unserer Zeit, No. 1 , pp. 56-61 , 2001 ) discloses general aspects in connection with insulin formulations. The German product specification of the medicinal product Berlinsulin® H Normal 3ml_ Pen discloses a formulation containing human insulin, metacresol, glycerol, water and optionally hydrochloric acid and sodium hydroxide for pH adjustment. The German product specification of the medicinal product Actrapid® discloses a formulation containing human insulin, zinc chloride, glycerol, metacresol, water and optionally sodium hydroxide and hydrochloric acid for pH adjustment. The FDA label of the medicinal product Lantus® discloses a formulation containing insulin glargine, zinc, m-cresol, glycerol 85 %, polysorbate 20 and water for injection, wherein the pH is adjusted to approximately 4 by addition of aqueous solutions of hydrochloric acid and/or sodium hydroxide. The FDA label of the medicinal product Humalog® discloses a formulation containing insulin lispro, glycerin, dibasic sodium phosphate, metacresol, zinc oxide, phenol and water for injection, wherein the pH is adjusted to 7.0-7.8 by addition of aqueous solutions of hydrochlorid acid and/or sodium hydroxide.
The solubility of insulin, insulin analogues and/or insulin derivatives in aqueous media depends on the pH value. For example, the lowest solubility is shown close to the isoelectric point which for human insulin is around pH 5.3 and 5.4. Very good solubility can be observed at pH values below 4 and above 7. However, insulin suffers from degradation at strong acidic conditions and strong alkaline conditions. Therefore, most of the medicinal products containing insulin, insulin analogues and/or insulin derivatives have a pH value in the range of 7.2 to 7.4 and mostly buffering agents are used to achieve and maintain the pH within this range. It has now surprisingly been found that an alternative aqueous pharmaceutical formulation of insulin aspart having less than 0.17 mg/ml_ sodium chloride shows an excellent chemical and physical stability which qualifies this aqueous pharmaceutical formulation as a medicinal product having a defined shelf life.
SUMMARY OF THE INVENTION
One embodiment of the present invention relates to a pharmaceutical formulation comprising (a), insulin aspart; and
(b). Zn(ll); and
(d). optionally protamine;
wherein the pharmaceutical formulation contains no or less than 0.17 mg/ml_ sodium chloride.
In another embodiment, the pharmaceutical formulation according to the present invention consists of (a), insulin aspart; and (b). Zn(ll); and (c). sodium chloride; and (d). optionally protamine; and (e). metacresol; (f). phenol; and (g). disodium phosphate (Na2HP04); (h). glycerol; and (i). sodium hydroxide and/or hydrochloric acid for pH adjustment to a pH value in the range of from 6.0 to 9.0; and (j)- water. In another embodiment, the pharmaceutical formulation according to the present invention is an aqueous pharmaceutical formulation.
In another embodiment, the pharmaceutical formulation according to the present invention comprises at least one analogue and/or derivative of insulin which has or have an isoelectric point (lEP) from 4.0 to 6.0, from 4.5 to 6.0, from 4.5 to 5.5, from 5.0 to 5.5, from 5.0 to 5.2 or 5.1.
In another embodiment, the pharmaceutical formulation according to the present invention has a pH value in the range from 6.0 to 9.0, from 6.5 to 8.5, from 7.0 to 8.0, from 7.0 to 7.8, from 7.1 to 7.6, or 7.2, 7.3, 7.4 or 7.5, or 7.4.
In another embodiment, the pharmaceutical formulation according to the present invention comprises insulin aspart which is present in a concentration from 10 U/mL to 1000 U/mL, from 10 U/mL to 600 U/mL, from 10 U/mL to 300 U/mL, from 50 U/mL to 300 U/mL or 100 U/mL. In another embodiment, the pharmaceutical formulation according to the present invention comprises insulin aspart which is present in a concentration from 60 to 6000 nmol/mL, from 60 nmol/mL to 3600 nmol/mL, from 60 nmol/mL to 1800 nmol/mL, from 300 nmol/mL to 1800 nmol/mL or 600 nmol/mL.
In another embodiment, the pharmaceutical formulation according to the present invention comprises Zn(ll) which is present in a concentration from 0.0100 mg/mL to 0.0600 mg/mL, from 0.0150 mg/mL to 0.0500 mg/mL, from 0.0150 mg/mL to 0.0300 mg/mL, from 0.0150 mg/mL to 0.0200 mg/mL, from 0.0190 mg/mL to 0.0200 mg/mL, or 0.0196 mg/mL.
In another embodiment, the pharmaceutical formulation according to the present invention comprises zinc chloride (ZnCI2) or zinc oxide (ZnO) or zinc acetate (anhydrous: C4H604Zn or dehydrate C4H604Zn x 2H20). In another embodiment, the pharmaceutical formulation according to the present invention comprises Zn(ll) which is present in a concentration from 0.0100 mg/100 U to 0.0600 mg/100 U, from 0.0150 mg/100 U to 0.0500 mg/100 U, from 0.0150 mg/100 U to 0.0300 mg/100 U, from 0.0150 mg/100 U to 0.0200 mg/100 U, from 0.0190 mg/100 U to 0.0200 mg/100 U, or 0.0196 mg/100 U.
In another embodiment, the pharmaceutical formulation according to the present invention comprises sodium chloride which is present in a concentration of less than 0.17 mg/mL, from 0 mg/mL to 0.17 mg/mL, from 0 mg/mL to 0.10 mg/mL, from 0 mg/mL to 0.05 mg/mL, or from 0 mg/mL to 0.01 mg/mL.
In another embodiment, the pharmaceutical formulation according to the present invention does not contain any sodium chloride.
In another embodiment, the pharmaceutical formulation according to the present invention is essentially free of sodium chloride.
In another embodiment, the pharmaceutical formulation according to the present invention further contains a buffering agent. In another embodiment, the pharmaceutical formulation according to the present invention comprises protamine or protamine sulfate which is present in a concentration from 0.10, 0.15, 0.20, 0.25, 0.30, 0.32, 0.35, 0.40, 0.45 or 0.5 mg/mL. In another embodiment, the pharmaceutical formulation according to the present invention comprises a stabilizing agent, which is in one embodiment a surfactant, a polyoxyethylene derivative of sorbitan monolaurate (e.g. polysorbate 20), a polyethoxylethylene derivate of oleic acid (e.g. polysorbate 80), poloxamer (which is a polyoxyethylene-polyoxypropylene copolymer), or polysorbate 20 or polysorbate 80 or mixtures thereof. In another embodiment, the stabilizing agent, in one embodiment the surfactant, the polyoxyethylene derivative of sorbitan monolaurate (e.g. polysorbate 20), the polyethoxylethylene derivate of oleic acid (e.g. polysorbate 80), poloxamer (which is a polyoxyethylene-polyoxypropylene copolymer), or polysorbate 20 or polysorbate 80 or mixtures thereof are/is present in a concentration from 0.01 to 0.05 mg/mLor in a concentration of 0.010 mg/mL, 0.015 mg/mL, 0.020 mg/mL, 0.025 mg/mL, 0.03 mg/mL, or 0.02 mg/mL.
In another embodiment, the pharmaceutical formulation according to the present invention comprises insulin aspart and one or more further analogues and/or derivatives of insulin, including the following combinations
(i). insulin aspart and one or more further analogue of insulin;
(ii). insulin aspart and one or more derivative of insulin;
(iii). Insulin aspart and one or more analogues of insulin and in addition one or more derivative of insulin.
In each combination, the one or more analogue of insulin may be a fast acting insulin or a long acting insulin. In each combination, the one or more derivative of insulin may be a fast acting insulin or a long acting insulin. In another embodiment, the fast acting insulin is selected from the group comprising insulin aspart, insulin lispro and/or insulin glulisine. In another embodiment, the long acting insulin is selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec. In another embodiment, pharmaceutical formulation according to the present invention comprises insulin aspart and a long acting insulin (either an analogue or a derivative of insulin), wherein the long acting insulin is in one embodiment selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec.
In another embodiment, the pharmaceutical formulation according to the present invention comprises one or more further active pharmaceutical ingredients. In one embodiment, the further active pharmaceutical ingredient is an antidiabetic agent. In another embodiment, the pharmaceutical formulation according to the present invention comprises one or more antidiabetic agents as further active pharmaceutical ingredients selected from the group comprising: GLP-1 receptor agonists, dual GLP-1 receptor/glucagon receptor agonists, human FGF-21 , FGF-21 analogues, FGF-21 derivatives, insulins, human insulin, analogues of insulin, and derivatives of insulin. In another embodiment, the pharmaceutical formulation according to the present invention comprises one or more further active pharmaceutical ingredients selected from the group comprising: insulin and insulin derivatives, GLP-1 , GLP-1 analogues and GLP-1 receptor agonists, polymer bound GLP-1 and GLP-1 analogues, dual GLP1/GIP agonists, dual GLP1/Glucagon receptor agonists, PYY3-36 or analogues thereof, pancreatic polypeptide or analogues thereof, glucagon receptor agonists or antagonists, GIP receptor agonists or antagonists, ghrelin antagonists or inverse agonists, Xenin and analogues thereof, DDP-IV inhibitors, SGLT2 inhibitors, dual SGLT2 / SGLT1 inhibitors, biguanides thiazolidinediones, dual PPAR agonists, sulfonylureas, meglitinides, alpha-glucosidase inhibitors, amylin and amylin analogues, GPR1 19 agonists, GPR40 agonists, GPR120 agonists, GPR142 agonists, systemic or low-absorbable TGR5 agonists, Cycloset, inhibitors of 1 1-beta-HSD, activators of glucokinase, inhibitors of DGAT, inhibitors of protein tyrosinephosphatase 1 , inhibitors of glucose-6-phosphatase, inhibitors of fructose-1 ,6- bisphosphatase, inhibitors of glycogen phosphorylase, inhibitors of phosphoenol pyruvate carboxykinase, inhibitors of glycogen synthase kinase, inhibitors of pyruvate dehydrogenase kinase, alpha2-antagonists, CCR-2 antagonists, modulators of glucose transporter-4, somatostatin receptor 3 agonists, HMG-CoA-reductase inhibitors, fibrates, nicotinic acid and the derivatives thereof, nicotinic acid receptor 1 agonists, PPAR-alpha, gamma or alpha/gamma) agonists or modulators, PPAR-delta agonists, ACAT inhibitors, cholesterol absorption inhibitors, bile acid- binding substances, IBAT inhibitors, MTP inhibitors, modulators of PCSK9, LDL receptor up- regulators by liver selective thyroid hormone receptor β agonists, HDL-raising compounds, lipid metabolism modulators, PLA2 inhibitors , ApoA-l enhancers, cholesterol synthesis inhibitors, lipid metabolism modulators, omega-3 fatty acids and derivatives thereof, active substances for the treatment of obesity, such as
sibutramine, tesofensine, orlistat, CB-1 receptor antagonists, MCH-1 antagonists, MC4 receptor agonists and partial agonists, NPY5 or NPY2 antagonists, NPY4 agonists, beta-3-agonists, leptin or leptin mimetics, agonists of the 5HT2c receptor, or the combinations of bupropione/naltrexone (CONTRAVE), bupropione/zonisamide (EMPATIC), bupropione/phentermine or
pramlintide/metreleptin, QNEXA (Phentermine+ topiramate), lipase inhibitors, angiogenesis inhibitors, H3 antagonists, AgRP inhibitors, triple monoamine uptake inhibitors (norepinephrine and acetylcholine), MetAP2 inhibitors, nasal formulation of the calcium channel blocker diltiazem, antisense oligonucleotides against production of fibroblast growth factor receptor 4, prohibitin targeting peptide-1 , drugs for influencing high blood pressure, chronic heart failure or atherosclerosis, such as angiotensin II receptor antagonists, ACE inhibitors, ECE inhibitors, diuretics, beta-blockers, calcium antagonists, centrally acting hypertensives, antagonists of the alpha-2-adrenergic receptor, inhibitors of neutral endopeptidase, thrombocyte aggregation inhibitors.
In another embodiment, the pharmaceutical formulation according to the present invention consists of (a). 3.5 mg/mL insulin aspart; and (b). 1.72 mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). 0.0196 mg/mL Zn(ll); and (e).1.88 mg/mL Na2HP04 x 7 H20 ; (f). 17.88 mg/mL glycerol; and (g). sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4 and (h). water.
In another embodiment, the pharmaceutical formulation according to the present invention consists of (a). 3.5 mg/mL insulin aspart; and (b). 1.72 mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). 0.0196 mg/mL Zn(ll); and (e).1.88 mg/mL Na2HP04 x 7 H20 ; (f). 17.88 mg/mL glycerol; (g). from 0.1 mg/mL to 0.5 mg/mL protamine sulfate; and (h). sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4 and (i). water.
In another embodiment, the pharmaceutical formulation according to the present invention consists of (a). 3.5 mg/mL insulin aspart; and (b). 1.72 mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). 0.0196 mg/mL Zn(ll); and (e).1.88 mg/mL Na2HP04 x 7 H20 ; (f). 17.88 mg/mL glycerol; (g). 0.1 or 0.15 or 0.2 or 0.25 or 0.3 or 0.32 or 0.35 or 0.4 or 0.45 or 0.5 mg/mL protamine sulfate; (h). less than 0.17 mg/mL sodium chloride; and (j)- sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4 and (i). water. The present invention also provides to a pharmaceutical formulation according to the present invention for use in the treatment of diabetes mellitus, hyperglycemia and/or for use in lowering blood glucose levels.
The present invention also provides to a process for preparing the pharmaceutical formulation according to the present invention, wherein the components are mixed together in the form of a solution or suspension, the desired pH is adjusted and the mixture is made up to the final volume with water.
The present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention and of a medical device. In one embodiment, the medical device is selected from the group comprising: syringe, insulin injection system, insulin infusion system, insulin pump, insulin pen injection device.
The present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device. In one embodiment, the medical device is selected from the group comprising: syringe, insulin injection system, insulin infusion system, insulin pump, insulin pen injection device. The present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the further active
pharmaceutical ingredient is an antidiabetic agent. The present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the further active
pharmaceutical ingredient is an antidiabetic agent selected from the group comprising: GLP-1 receptor agonists, dual GLP-1 receptor/glucagon receptor agonists, human FGF-21 , FGF-21 analogues, FGF-21 derivatives, insulins, human insulin, analogues of insulin, and derivatives of insulin. In anotherembodiment, the pharmaceutical formulation according to the present invention comprises one or more further active pharmaceutical ingredients selected from the group comprising: insulin and insulin derivatives, GLP-1 , GLP-1 analogues and GLP-1 receptor agonists, polymer bound GLP-1 and GLP-1 analogues, dual GLP1/GIP agonists, dual GLP1/Glucagon receptor agonists, PYY3-36 or analogues thereof, pancreatic polypeptide or analogues thereof, glucagon receptor agonists or antagonists, GIP receptor agonists or antagonists, ghrelin antagonists or inverse agonists, Xenin and analogues thereof, DDP-IV inhibitors, SGLT2 inhibitors, dual SGLT2 / SGLT1 inhibitors, biguanides thiazolidinediones, dual PPAR agonists, sulfonylureas, meglitinides, alpha-glucosidase inhibitors, amylin and amylin analogues, GPR1 19 agonists, GPR40 agonists, GPR120 agonists, GPR142 agonists, systemic or low-absorbable TGR5 agonists, Cycloset, inhibitors of 1 1-beta-HSD, activators of glucokinase, inhibitors of DGAT, inhibitors of protein tyrosinephosphatase 1 , inhibitors of glucose-6-phosphatase, inhibitors of fructose-1 ,6- bisphosphatase, inhibitors of glycogen phosphorylase, inhibitors of phosphoenol pyruvate carboxykinase, inhibitors of glycogen synthase kinase, inhibitors of pyruvate dehydrogenase kinase, alpha2-antagonists, CCR-2 antagonists, modulators of glucose transporter-4, somatostatin receptor 3 agonists, HMG-CoA-reductase inhibitors, fibrates, nicotinic acid and the derivatives thereof, nicotinic acid receptor 1 agonists, PPAR-alpha, gamma or alpha/gamma) agonists or modulators, PPAR-delta agonists, ACAT inhibitors, cholesterol absorption inhibitors, bile acid- binding substances, IBAT inhibitors, MTP inhibitors, modulators of PCSK9, LDL receptor up- regulators by liver selective thyroid hormone receptor β agonists, HDL-raising compounds, lipid metabolism modulators, PLA2 inhibitors , ApoA-l enhancers, cholesterol synthesis inhibitors, lipid metabolism modulators, omega-3 fatty acids and derivatives thereof, active substances for the treatment of obesity, such as sibutramine, tesofensine, orlistat, CB-1 receptor antagonists, MCH-1 antagonists, MC4 receptor agonists and partial agonists, NPY5 or NPY2 antagonists, NPY4 agonists, beta-3-agonists, leptin or leptin mimetics, agonists of the 5HT2c receptor, or the combinations of bupropione/naltrexone (CONTRAVE), bupropione/zonisamide (EMPATIC), bupropione/phentermine or pramlintide/metreleptin, QNEXA (Phentermine+ topiramate), lipase inhibitors, angiogenesis inhibitors, H3 antagonists, AgRP inhibitors, triple monoamine uptake inhibitors (norepinephrine and acetylcholine), MetAP2 inhibitors, nasal formulation of the calcium channel blocker diltiazem, antisense oligonucleotides against production of fibroblast growth factor receptor 4, prohibitin targeting peptide-1 , drugs for influencing high blood pressure, chronic heart failure or atherosclerosis, such as angiotensin II receptor antagonists, ACE inhibitors, ECE inhibitors, diuretics, beta-blockers, calcium antagonists, centrally acting hypertensives, antagonists of the alpha-2-adrenergic receptor, inhibitors of neutral endopeptidase, thrombocyte aggregation inhibitors.
The present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the kit comprises more than one analogue and/or derivative of insulin, wherein one analogue and/or derivative of insulin is a fast acting insulin and one analogue and/or derivative of insulin is a long acting insulin. In one embodiment, the fast acting insulin is selected from the group comprising insulin aspart, insulin lispro and/or insulin glulisine and wherein the long acting insulin is selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec.
The present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device for use in the treatment of diabetes mellitus, hyperglycemia and/or for use in lowering blood glucose levels. The present invention also relates to a kit comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the further active pharmaceutical ingredient is an analogue or derivative of insulin selected from the group of fast acting insulin and long acting insulin.
The present invention also relates to a kit comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the further active pharmaceutical ingredient is an analogue or derivative of insulin selected from the group of fast acting insulin and long acting insulin and wherein the fast acting insulin is selected from the group comprising insulin aspart, insulin lispro and/or insulin glulisine and wherein the long acting insulin is selected from the group comprising insulin glargin, insulin detemir and/or insulin degludec. In anotherembodiment, the present invention also relates to a kit or combination comprising separate packages of the pharmaceutical formulation according to the present invention, of at least one further active pharmaceutical ingredient and optionally of a medical device, wherein the pharmaceutical formulation according to the present invention and the further active pharmaceutical ingredient, in one embodiment an antidiabetic agent, are administered continuously, separately, sequentially and/or stepwise.
The present invention also relates to the use of a medical device for administering the
pharmaceutical formulation to an animal and/or human. In another embodiment, the medical device is selected from the group comprising: syringe, insulin injection system, insulin infusion system, insulin pump, insulin pen injection device
DETAILED DESCRIPTION
As used herein, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to a fill materiel containing "a carrier" includes one or more carriers, reference to "an additive" includes reference to one or more of such additives.
As used herein, the term "active pharmaceutical ingredient" (API) includes any pharmaceutically active chemical or biological compound and any pharmaceutically acceptable salt thereof and any mixture thereof, that provides some pharmacologic effect and is used for treating or preventing a condition. Exemplary pharmaceutically acceptable salts include hydrochloric, sulfuric, nitric, phosphoric, hydrobromic, maleric, malic, ascorbic, citric, tartaric, pamoic, lauric, stearic, palmitic, oleic, myristic, lauryl sulfuric, naphthalinesulfonic, linoleic, linolenic acid, and the like. As used herein, the terms "active pharmaceutical ingredient", "drug", "active agent", "active ingredient", "active substance" and "drug" are meant to be synonyms, i.e., have identical meaning.
In a one embodiment the active pharmaceutical ingredient is an antidiabetic agent. Examples of antidiabetic agents are found in the Rote Liste 2012, chapter 12. Examples of antidiabetic agents include but are not Imitied to (a) insulin, insulin analogues and insulin derivatives, (b) glucagon-like- peptide 1 (GLP-1 ) and its analogues and receptor agonists, (c) dual GLP-1/GIP agonists, and (d) dual GLP-1 /glucagon receptor agonists, as described in detail next.
(a) . Insulin, insulin analogues and insulin derivatives
Examples of insulin, insulin analogues, and insulin derivatives include but are not limited to insulin glargine (Lantus®), insulin glulisine (Apidra®), insulin detemir (Levemir®), insulin lispro (Humalog® / Liprolog®), insulin degludec (Tresiba®), insulin aspart (NovoLog® / NovoRapid®), basal insulin and analogues (e.g. LY2605541 , LY2963016), PEGylated insulin lispro, Humulin®, Linjeta®, SuliXen®, NN1045, insulin plus Symlin®, fast-acting and short-acting insulins (e.g. Linjeta®, PH20 insulin, NN1218, HinsBet®), oral, inhalable, transdermal and sublingual insulins (e.g. Exubera®, Nasulin®, Afrezza®, insulin tregopil, TPM -02/lnsulin, Capsulin®, Oral-lyn®, Cobalamin® oral insulin, ORMD- 0801 , NN1953, VIAtab®). Additionally included are also those insulin derivatives which are bonded to albumin or another protein by a bifunctional linker.
(b) . Glucagon-like-peptide 1 (GLP-1 ), GLP-1 analogues and GLP-1 receptor agonists
Examples: GLP-1 , GLP-1 analogues and GLP-1 receptor agonists include but are not limited to lixisenatide (Lyxumia®), exenatide / exendin-4 (Byetta® / Bydureon® / ITCA 650, liraglutide / Victoza®), semaglutide, taspoglutide, albiglutide, dulaglutide, rExendin-4, CJC-1 134-PC, PB-1023, TTP-054, HM-1 1260C, CM-3, GLP-1 Eligen, ORMD-0901 , NN9924, Nodexen, Viador-GLP-1 , CVX- 096, ZYOG-1 , ZYD-1 , MAR-701 , ZP-2929, ZP-3022, CAM-2036, DA-15864, ARI-2651 , ARI-2255, exenatide-XTEN and glucagon-XTEN, AMX-8089+VRS-859 and polymer bound GLP-1 and GLP-1 analogues.
(c). Dual GLP-1 /glucose-dependent insulinotropic peptides (GIP) agonists
Examples of dual GLP-1/GIP agonists include but are not limited to MAR701 , MAR-709, BHM081 / BHM089 / BHM098). (d). Dual GLP-1 /glucagon receptor agonists
Examples of dual GLP-1/glucagon receptor agonists include but are not limited to OAP-189 (PF- 05212389, TKS-1225), TT-401/402, ZP2929, LAPS-HMOXM25, MOD-6030).
Other suitable active pharmaceutical ingredients which may be included in the pharmaceutical formulations of the invention include but are not limited to the following:
Further gastrointestinal peptides such as peptide YY 3-36 (PYY3-36) or analogues thereof and pancreatic polypeptide (PP) or analogues thereof.
Glucagon receptor agonists or antagonists, GIP receptor agonists or antagonists, ghrelin antagonists or inverse agonists and xenin and analogues thereof.
Dipeptidyl peptidase-IV (DPP-4) inhibitors, for example: alogliptin / Nesina®, linagliptin / BI-1356 / Ondero® / Trajenta® / Tradjenta® / Trayenta®, saxagliptin / Onglyza®, sitagliptin / Januvia® / Xelevia® / Tesavel®, sitagliptin + metformin / Janumet® / Velmetia®, aildagliptin, anagliptin, aemigliptin, tenegliptin, melogliptin, trelagliptin, DA-1229, MK-3102, KM-223, KRP-104 and Ari-2243.
Sodium-dependent glucose transporter 2 (SGLT2) inhibitors, for example: canagliflozin, dapagliflozin, remogliflozin, sergliflozin, empagliflozin, ipragliflozin, tofogliflozin (RO-4998452), luseogliflozin, LX-4211 , ertugliflozin (PF-04971729), EGT-0001442 and DSP-3235.
Dual SGLT2 / SGLT1 inhibitors.
Biguanides (e.g. metformin, buformin, phenformin), thiazolidinediones (e.g. pioglitazone, rivoglitazone, rosiglitazone, troglitazone), dual PPAR agonists (e.g. aleglitazar, muraglitazar, tesaglitazar), sulfonylureas (e.g. tolbutamide, glibenclamide, glimepiride / Amaryl®, glipizide), meglitinides (e.g. nateglinide, repaglinide, mitiglinide), alpha-glucosidase inhibitors (e.g. acarbose, miglitol, voglibose), amylin and amylin analogues (e.g. pramlintide / Symlin®).
G-protein coupled receptor 119 (GPR1 19) agonists (e.g. GSK-1292263, PSN-821 , MBX-2982, APD-597, ARRY-981 ).
GPR40 agonists (e.g. TAK-875, TUG-424, P-1736, JTT-851 , GW9508).
GPR120 agonists and GPR142 agonists.
Systemic or low-absorbable TGR5 (GPBAR1 = G-protein-coupled bile acid receptor 1 ) agonists (e.g. INT-777, XL-475, SB756050).
Bromocriptine / Cycloset®, inhibitors of 1 1 -beta-hydroxysteroid dehydrogenase (1 1 -beta-HSD) (e.g. LY2523199, BMS770767, RG-4929, BMS816336, AZD-8329, HSD-016, BI-135585), activators of glucokinase (e.g. PF-04991532, TTP-399, GK1 -399, ARRY-403 (AMG-151 ), TAK-329, ZYGK1 ), inhibitors of diacylglycerol O-acyltransferase (DGAT) (e.g. pradigastat (LCQ-908), LCQ-908), inhibitors of protein tyrosinephosphatase 1 (e.g. trodusquemine), inhibitors of glucose-6- phosphatase, inhibitors of fructose-1 ,6-bisphosphatase, inhibitors of glycogen phosphorylase, inhibitors of phosphoenol pyruvate carboxykinase, inhibitors of glycogen synthase kinase, inhibitors of pyruvate dehydrogenase kinase, alpha2 adrenergic receptor antagonists, C-C chemokine receptor type 2 (CCR-2) antagonists, modulators of glucose transporter-4 and somatostatin receptor 3 agonists (e.g. MK^1256).
One or more lipid lowering agents are also suitable as active pharmaceutical ingredients, such as for example: 3-hydroxy-3-methylglutaryl-coenzym-A-reductase (HMG-CoA-reductase) inhibitors (e.g. simvastatin, atorvastatin, rosuvastatin), fibrates (e.g. bezafibrate, fenofibrate), nicotinic acid and derivatives thereof (e.g. niacin, including slow release formulations of niacin), nicotinic acid receptor 1 agonists (e.g. GSK-256073), peroxisome proliferator-activated receptors (PPAR-)(alpha, gamma or alpha/gamma) agonists or modulators (e.g. aleglitazar), PPAR-delta agonists, acetyl- CoA-acetyltransferase (ACAT) inhibitors (e.g. avasimibe), cholesterol absorption inhibitors (e.g. ezetimibe), bile acid-binding substances (e.g. cholestyramine, colesevelam), ileal bile acid transport inhibitors (IBAT) (e.g. GSK-2330672), microsomal triglyceride transfer protein (MTP) inhibitors (e.g. lomitapide (AEGR-733), SLx-4090, granotapide), modulators of proprotein convertase
subtilisin/kexin type 9 (PCSK9) (e.g. REGN727/SAR236553, AMG-145, LGT-209, PF-04950615, MPSK3169A, LY3015014, ALD-306, ALN-PCS, BMS-962476, SPC5001 , ISIS-394814, 1 B20, LGT-210, 1 D05, BMS-PCSK9Rx-2, SX-PCK9, RG7652), LDL receptor up-regulators, for example liver selective thyroid hormone receptor beta agonists (e.g. eprotirome (KB-21 15), MB0781 1 , sobetirome (QRX-431 ), VIA-3196, ZYT1 ), HDL-raising compounds such as: CETP inhibitors (e.g. torcetrapib, anacetrapib (MK0859), dalcetrapib, evacetrapib, JTT-302, DRL-17822, TA-8995, R- 1658, LY-2484595) or ABC1 regulators, lipid metabolism modulators (e.g. BMS-823778, TAP-301 , DRL-21994, DRL-21995), phospholipase A2 (PLA2) inhibitors (e.g. darapladib / Tyrisa®, varespladib, rilapladib), ApoA-l enhancers (e.g. RVX-208 , CER-001 , MDCO-216, CSL-1 12, VRX- HDL, VRX-1243, VIRxSYS), cholesterol synthesis inhibitors (e.g. ETC-1002) and lipid metabolism modulators (e.g. BMS-823778, TAP-301 , DRL-21994, DRL-21995) and omega-3 fatty acids and derivatives thereof (e.g. icosapent ethyl (AMR101 ), Epanova®, AKR-063, NKPL-66).
Other suitable active pharmaceutical ingredients ingredients which may be included in the pharmaceutical formulations include one or more active substances for the treatment of obesity, including but not limited to:
Sibutramine, tesofensine, orlistat, cannabinoid receptor 1 (CB1 ) antagonists (e.g. TM-38837), melanin-concentrating hormone (MCH-1 ) antagonists (e.g. BMS-830216, ALB-127158(a)), MC4 receptor agonists and partial agonists (e.g. AZD-2820, RM-493 ), neuropeptide Y5 (NPY5) or NPY2 antagonists (e.g. velneperit, S-234462), NPY4 agonists (e.g. PP-1420), beta-3-adrenergic receptor agonists, leptin or leptin mimetics, agonists of the 5-hydroxytryptamine 2c (5HT2c) receptor (e.g. lorcaserin), or the combinations of bupropione/naltrexone (Contrave®),
bupropione/zonisamide (Empatic®), bupropione/phentermine or pramlintide/metreleptin, phentermine/topiramate (Qsymia®), lipase inhibitors (e.g. cetilistat / Cametor®), angiogenesis inhibitors (e.g. ALS-L1023), histamine H3 antagonists (e.g. HPP-404), AgRP (agouti related protein) inhibitors (e.g. TTP-435), triple monoamine uptake inhibitors (dopamine, norepinephrine and serotonin reuptake) (e.g. tesofensine), methionine aminopeptidase 2 (MetAP2) inhibitors (e.g. beloranib), nasal formulations of the calcium channel blocker diltiazem (e.g. CP-404) and antisense oligonucleotides against production of fibroblast growth factor receptor 4 (FGFR4) (e.g. ISIS- FGFR4Rx) or prohibitin targeting peptide-1 (e.g. Adipotide®).
Further suitable active pharmaceutical ingredients which may be included in the pharmaceutical formulations include but are not limited to:
Angiotensin II receptor antagonists (e.g. telmisartan, candesartan, valsartan, losartan, eprosartan, irbesartan, olmesartan, tasosartan, azilsartan), angiotensin converting enzyme (ACE) inhibitors, endothelin converting enzyme (ECE) inhibitors, diuretics, beta-blockers, calcium antagonists, centrally acting hypertensives, antagonists of the alpha-2-adrenergic receptor, inhibitors of neutral endopeptidase, thrombocyte aggregation inhibitors and others or combinations thereof are suitable. As used herein, the terms ..analogue of insulin" and "insulin analogue" refers to a polypeptide which has a molecular structure which formally can be derived from the structure of a naturally occurring insulin, for example that of human insulin, by deleting and/or exchanging at least one amino acid residue occurring in the naturally occurring insulin and/or adding at least one amino acid residue. The added and/or exchanged amino acid residue can either be codable amino acid residues or other naturally occurring residues or purely synthetic amino acid residues. Examples of analogues of insulin include, but are not limited to, the following: (i). 'Insulin aspart' is created through recombinant DNA technology so that the amino acid B28 in human insulin (i.e. the amino acid no. 28 in the B chain of human insulin), which is proline, is replaced by aspartic acid; (ii). 'Insulin lispro' is created through recombinant DNA technology so that the penultimate lysine and proline residues on the C-terminal end of the B-chain of human insulin are reversed (human insulin: ProB28LysB29; insulin lispro: LysB28ProB29); (iii). 'Insulin glulisine' differs from human insulin in that the amino acid asparagine at position B3 is replaced by lysine and the lysine in position B29 is replaced by glutamic acid; (iv). "Insulin glargine" differs from human insulin in that the asparagine at position A21 is replaced by glycine and the B chain is extended at the carboxy terminal by two arginines. As used herein, the term "aqueous" refers to a solution in which the solvent is water and/or to a suspension in which the external phase is water and/or to an emulsion in which the dispersed or continuous phase is water. As used herein, the term "buffering agent" refers to a weak acid or base used to maintain the acidity (pH) of a solution, a suspension and/or an emulsion near a chosen value after the addition of another acid or base. The function of a buffering agent is to prevent a rapid change in the pH value when acids or bases are added to the solution. In an aqueous solution, suspension and/or emulsion, a buffering agent is present in a mixture of a weak acid and its conjugate base or a in a mixture of a weak base and its conjugated acid. Examples of buffering agents include, but are not limited to, the following: sodium bicarbonate; acetic acid or acetate salts (e.g. sodium acetate, zinc acetate); boric acid or boric salts; N-cyclohexyl-2-aminoethanesulfonic acid (CHES) or salts thereof; 3-[[1 ,3-dihydroxy-2-(hydroxymethyl)propan-2-yl]amino]propane-1-sulfonic acid (TAPS) or salts thereof; 2-(N-morpholino)ethanesulfonic acid (MES) and salts therof; piperazine-Ν,Ν -bis(2- ethanesulfonic acid (PIPES) and salts therof; N-(2-acetamido)-2-aminoethane-sulfonic acid (ACES) and salts therof; cholamine chloride; BES; 2-[[1 ,3-dihydroxy-2-(hydroxymethyl)-propan-2- yl]amino]ethanesulfonic acid (TES) and salts therof; 2-[4-(2-hydroxyethyl)piperazin-1 - yljethanesulfonic acid (HEPES) and salts therof; acetamidoglycine; N-(2-hydroxy-1 , 1-bis(hydroxyl- methyl)ethyl)glycine (tricine); glycinamide; 2-(bis(2-hydroxyethyl)amino)acetic acid (bicine) and salts therof; propionate salts; 3-[[1 ,3-dihydroxy-2-(hydroxymethyl)propan-2-yl]-amino]-2-hydroxy- propane-1 -sulfonic acid (TAPSO) and salts therof; 3-morpholinopropane-1 -sulfonic acid (MOPS) and salts therof; saline-sodium citrate (SSC) buffer; 2-amino-2-hydroxymethyl-propane-1 ,3-diol (synonyms: TRIS, trisamine, THAM, tromethamine, trometamol, tromethane); citric acid or citrate salts (e.g. sodium citrate); trisodium phosphate, disodium hydrogen phosphate, sodium dihydrogen phosphate, tripotassium phosphate, dipotassium phosphate, monopotassium phosphate and/or any other buffering agent containing phosphate.
Amino acids (having free basic or acidic functional groups, e.g. methionin, arginine) or peptides (having free basic or acidic functional groups) may also be used as buffering agent. As used herein, the term "buffering agent" also comprises amino acids, peptides and proteins. As insulin analogues and/or insulin derivatives and/or protamine are peptides or derivatives of peptides (i.e. both contain amino acids having free basic or acidic functional groups), they may also have a certain buffering capacity, i.e. are also to be considered as buffering agent.
As used herein, the term "fast acting insulin" or "short acting insulin" refers to insulin analogues and/or insulin derivatives, wherein the insulin-mediated effect begins within 5 to 15 minutes and continues to be active for 3 to 4 hours. Examples of fast acting insulins include, but are not limited to, the following: (i). insulin aspart; (ii). insulin lispro and (iii). insulin glulisine.
As used throughout the description and the claims of this specification, the word "comprise" and variations of the word, such as "comprising" and "comprises" is not intended to exclude other additives, components, integers or steps.
As used herein, the terms ..derivative of insulin" and "insulin derivative" refers to a polypeptide which has a molecular structure which formally can be derived from the structure of a naturally occurring insulin, for example that of human insulin, in which one or more organic substituent (e.g. a fatty acid) is bound to one or more of the amino acids. Optionally, one or more amino acids occurring in the naturally occurring insulin may have been deleted and/or replaced by other amino acids, including non-codeable amino acids, or amino acids, including non-codeable, have been added to the naturally occurring insulin. Examples of derivatives of insulin include, but are not limited to, the following: (i). 'Insulin detemir' which differs from human insulin in that the C-terminal threonine in position B30 is removed and a fatty acid residue (myristic acid) is attached to the epsilon-amino function of the lysine in position B29. (ii). 'Insulin degludec' which differs from human insulin in that the last amino acid is deleted from the B-chain and by the addition of a glutamyl link from LysB29 to a hexadecandioic acid.
As used herein, the term "FGF-21 " means "fibroblast growth factor 21 ". FGF-21 compounds may be human FGF-21 , an analogue of FGF-21 (referred to "FGF-21 analogue") or a derivative of FGF-21 (referred to "FGF-21 derivative"). As used herein, the term "formulation" refers to a product comprising specified ingredients in predetermined amounts or proportions, as well as any product that results, directly or indirectly, from combining specified ingredients in specified amounts. In relation to pharmaceutical
formulations, this term encompasses a product comprising one or more active ingredients, and an optional carrier comprising inert ingredients, as well as any product that results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. In general, pharmaceutical formulations are prepared by uniformly bringing the active pharmaceutical ingredient (i.e. the analogue and/or derivative of insulin) into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. The pharmaceutical formulation includes enough of the active pharmaceutical ingredient to produce the desired effect upon the progress or condition of diseases. As used herein, the term "formulation" may refer to a solution as well as to a suspension or to an emulsion. As used herein, the terms "formulation" and "composition" are meant to be synonyms, i.e., have identical meaning. The pharmaceutical compositions are made following conventional techniques of pharmaceutical technology involving mixing, filling and dissolving the ingredients, as appropriate, to give the desired oral, parenteral, rectal, transdermal, or topical products.
As used herein, the term "GLP-1 receptor agonist" refers to compounds which have an agonistic activity at the glucagon-like peptide-1 receptor. Examples of GLP-1 receptor agonists include, but are not limited to, the following: exenatide / exendin-4, liraglutide, lixisenatide, dulaglutide, albiglutide, semaglutide, taspoglutide, rExendin-4, CJC-1 134-PC, PB-1023, TTP-054, HM-1 1260C, CM-3, GLP-1 Eligen, ORMD-0901 , NN9924, Nodexen, Viador-GLP-1 , CVX-096, ZYOG-1 , ZYD-1 , MAR-701 , ZP-2929, ZP-3022, CAM-2036, DA-15864, ARI-2651 , ARI-2255, exenati de-XT EN and glucagon-XTEN, AMX-8089+VRS-859 and polymer bound GLP-1 and GLP-1 analogues.
As used herein, the term "dual GLP-1 receptor/glucagon receptor agonist" refers to compounds which have agonistic activity at both the GLP-1 receptor and the glucacon receptor. Examples of dual GLP-1 receptor/glucagon receptor agonist include, but are not limited to, the following:
oxyntomodulin, MAR701 , MAR-709, and BHM081 / BHM089 / BHM098.
As used herein, the term "human insulin" refers to the human hormone whose structure and properties are well-known. Human insulin has two polypeptide chains (chains A and B) that are connected by disulphide bridges between cysteine residues, namely the A-chain and the B-chain. The A-chain is a 21 amino acid peptide and the B-chain is a 30 amino acid peptide, the two chains being connected by three disulphide bridges: one between the cysteins in position 6 and 1 1 of the A-chain; the second between the cysteine in position 7 of the A-chain and the cysteine in position 7 of the B-chain; and the third between the cysteine in position 20 of the A-chain and the cysteine in position 19 of the B-chain.
As used herein, the term "including" is used to mean "including but not limited to". "Including" and "including but not limited to" are used interchangeably.
As used herein, the term "isoelectric point" (pi, IEP) refers to the pH value at which a particular molecule carries no net electrical charge. The isoelectric point can be determined by using isoelectric focusing, which is a technique for separating different molecules by differences in their isoelectric point and which is well known in the art. It can also be calculated (see e.g. Levene and Simms, 'Calculation of isoelectric point' J. Biol. Chem., 1923, pp. 801 -813). As used herein, the term "kit" refers to a product (e.g. medicament, kit-of-parts) comprising one package or one or moreseparate packages of:
(i) . A pharmaceutical formulation containing an active pharmaceutical ingredient and at least one further active pharmaceutical ingredient and optionally a medical device. The at least one further active pharmaceutical ingredient may be present in said pharmaceutical formulation, i.e. the kit may comprise one or more packages, wherein each package comprises one pharmaceutical formulation which comprises two or more active pharmaceutical ingredients. The further active pharmaceutical ingredient may also be present in a further pharmaceutical formulation, i.e. the kit may comprise separate packages of two or more pharmaceutical formulations, wherein each pharmaceutical formulation contain one active pharmaceutical ingredient.
Or
(ii) . A pharmaceutical formulation containing an active pharmaceutical ingredient and medical device.
A kit may comprise one package only or may comprise one or more separate packages
For example, the kit may be a product (e.g. medicament) containing two or more vials each containing a defined pharmaceutical formulation, wherein each pharmaceutical formulation contains at least one active pharmaceutical ingredient. For example, the kit may comprise (i.) a vial containing a defined pharmaceutical formulation and (ii). further a tablet, capsule, powder or any other oral dosage form which contains at least one further active pharmaceutical ingredient. The kit may further comprise a package leaflet with instructions for how to administer the pharmaceutical formulation and the at least one further active pharmaceutical ingredient.
As used herein, the term "medical device" means an instrument, apparatus, implant, in vitro reagent or similar or related article that is used to diagnose, prevent, or treat a disease of other condition, and does not achieve its purpose through pharmacological action within or on the body. As used herein, a medical device may be a syringe, an insulin injection system, an insulin infusion system, an insulin pump or an insulin pen injection device. As used herein, a medical device may be mechanically or electromechanically driven. As used herein, unless specifically indicated otherwise, the conjunction "or" is used in the inclusive sense of "and/or" and not the exclusive sense of "either/or".
As used herein, the term "pH" and "pH value" refer to the decimal logarithm of the reciprocal of the hydrogen ion activity in a solution.
As used herein, the term "pharmaceutical" refers to the intended use in the medical diagnosis, cure, treatment and/or prevention of diseases. As used herein, the term "pharmaceutically acceptable" refers to physiologically well tolerated by a mammal or a human.
As used herein, the term "protamine" refers to a mixture of strongly basic peptides. It was originally isolated from the sperm of salmon and other species of fish but is now produced primarily recombinant through biotechnology. It contains more than two-thirds of L-arginine. As protamine contains amino acids having free basic side chains, it has a certain buffering capacity and is therefore considered to be a buffering agent. Protamine may be used as protamine sulfate or protamine hydrochloride. Concentrations, amounts, solubility in different liquid systems, particle size, wavelength, pH values, weight mass, molecular weight, percent and other numerical date may be expressed or presented herein in a range format. It is to be understood that such a range format is used merely for convenience and brevity and thus should be interpreted flexibly to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited.
As used herein, the term "long acting insulin" refers to insulin analogues and/or insulin derivatives, wherein the insulin-mediated effect begins within 0.5 to 2 hours and continues to be active, for about or more than 24 hours. Examples of fast acting insulins include, but are not limited to, the following: (i). insulin glargin; (ii). insulin detemir and (iii). insulin degludec.
As used herein, the term "stability" refers to the chemical and/or physical stability of active pharmaceutical ingredients, in particular of insulin analogues and/or derivatives. The purpose of stability testing is to provide evidence on how the quality of an active pharmaceutical ingredient or dosage form varies with time under the influence of a variety of environmental factors such as temperature, humidity, and light, and to establish a shelf life for the active pharmaceutical ingredient or dosage form and recommended storage conditions. Stability studies can include testing of those attributes of the active pharmaceutical ingredient that are susceptible to change during storage and are likely to influence quality, safety, and/or efficacy. The testing can cover, as appropriate, the physical, chemical, biological, and microbiological attributes, preservative content (e.g., antioxidant, antimicrobial preservative), and functionality tests (e.g. for a dose delivery system). Analytical procedures can be fully validated and stability indicating. In general, significant changes for an active pharmaceutical ingredient and/or dosage form with regard to stability are defined as:
• a 5% change in assay from its initial value; or failure to meet the acceptance criteria for potency when using biological or immunological procedures;
• any degradation products exceeding its acceptance criterion;
• failure to meet the acceptance criteria for appearance, physical attributes, and functionality test (e.g., color, phase, separation, resuspendibility, caking, hardness, dose delivery per actuation); however, some changes in physical attributes (e.g. softening of suppositories, melting of creams) may be expected under accelerated conditions;
and, as appropriate for the dosage form:
• failure to meet the acceptance criterion for pH; or
The significant changes may also be evaluated against established acceptance criteria prior to starting the evaluation of the stability.
Acceptance criteria can be derived from the monographs (e.g. monographs for the European Pharmacopeia, of the United States Pharmacopeia, of the British Pharmacopeia, or others), and from the analytical batches of the active pharmaceutical ingredient and medicinal product used in the preclinical and clinical studies. Acceptable limits should be proposed and justified, taking into account the levels observed in material used in preclinical and clinical studies. Product characteristics may be visual appearance, purity, color and clarity for solutions/suspensions, visible particulates in solutions, and pH. As a non-limiting example, suitable acceptance criteria for insulin aspart formulations are shown below:
Test item Acceptance criteria for clinical trials
Appearance of solution (visual)
- Clarity and degree of opalescence Monitoring
- Degree of coloration Monitoring
Assay insulin aspart units (HPLC) 90.0 insulin aspart units/mL to 1 10.0 insulin
Figure imgf000022_0001
nc n e ow μg per un s nsu n aspar
The acceptance criteria shown above are based on monographed acceptance limits (e.g. British Pharmacopoeia, Volume III, 2012 or Pharmacopoeial Forum, Volume 36(6), Nov-Dec 2010) and/or are derived from extensive experience in the development of insulin formulations.
As used herein, the term "treatment" refers to any treatment of a mammalian, for example human condition or disease, and includes: (1 ) inhibiting the disease or condition, i.e., arresting its development, (2) relieving the disease or condition, i.e., causing the condition to regress, or (3) stopping the symptoms of the disease.
As used herein, the unit of measurement„U" and/or Jnternational units" refers to the blood glucose lowering activity of insulin and is defined (according to the World Health Organization, WHO) as follows: 1 U corresponds to the amount of highly purified insulin (as defined by the WHO) which is sufficient to lower the blood glucose level of a rabbit (having a body weight of 2 - 2.5 kg) to 50 mg / 100 ml. within 1 hour and to 40 mg / 100 ml. within 2 hours. For human insulin, 1 U corresponds to approximately 35 μg (Lill, Pharmazie in unserer Zeit, No. 1 , pp. 56-61 , 2001 ). For insulin aspart, 100 U correspond to 3.5 mg (product information NovoRapid®). For insulin lispro, 100 U correspond to 3.5 mg (product information Humalog®). For insulin glulisine, 100 U correspond to 3.49 mg (product information Apidra® cartridges). For insulin determir, 100 U correspond to 14.2 mg (product information Levemir®). For insulin glargin, 100 U correspond to 3.64 mg (product information Lantus®). Further embodiments of the present invention include the following:
In one aspect, the invention provides a pharmaceutical formulation comprising (a), insulin aspart; and (b). Zn(ll); and (c). optionally protamine; wherein the pharmaceutical formulation contains less than 0.17 mg/ml_ sodium chloride.
In one aspect, the pharmaceutical formulation of the invention is an aqueous pharmaceutical formulation. In one aspect, the pharmaceutical formulation of the invention has a pH value in the range from 6.0 to 9.0.
In one aspect, the pharmaceutical formulation of the invention has a pH value in the range from 7.0 to 7.8.
In one aspect, the pharmaceutical formulation of the invention comprises insulin aspart which is present in a concentration from 10 U/mL to 1000 U/mL.
In one aspect, the pharmaceutical formulation of the invention comprises Zn(ll) which is present in a concentration from 0.0100 to 0.0600 mg / 100 U of insulin aspart.
In one aspect, the pharmaceutical formulation of the invention is essentially free of sodium chloride.
In one aspect, the pharmaceutical formulation of the invention comprises protamine which is present in a concentration from 0.1 to 0.5 mg/ml_.
In one aspect, the pharmaceutical formulation of the invention further contains a buffering agent.
In one aspect, the pharmaceutical formulation of the invention comprises one or more further active pharmaceutical ingredients.
In one aspect, the pharmaceutical formulation of the invention comprises a further active pharmaceutical ingredient which is an antidiabetic agent. In one aspect, the pharmaceutical formulation of the invention comprises a further active pharmaceutical ingredient which is an antidiabetic agent selected from the group consisting of: (a), a GLP-1 receptor agonist; (b). a dual GLP-1 receptor/glucagon receptor agonist; (c). human FGF-21 ; (d). a FGF-21 analogue; (e). a FGF-21 derivative; (f). insulin; (g). human insulin;
(h). an analogue of insulin; and (i). a derivative of insulin.
In one aspect, the pharmaceutical formulation of the invention further comprises a fast acting analogue and/or derivative of insulin or a long acting analogue and/or derivative of insulin. In one aspect, the pharmaceutical formulation of the invention comprises a fast acting analogue and/or derivative of insulin or a long acting analogue and/or derivative of insulin, wherein the fast acting insulin is one or more insulin selected from the group consisting of insulin lispro and insulin glulisine and wherein the long acting insulin is one or more insulin selected from the group consisting of insulin detemir, insulin glargin and insulin degludec.
In one aspect, the pharmaceutical formulation of the invention consists of: (a). 3.5 mg/ml_ insulin aspart; (b). 0.0196 mg/mL Zn(ll) ; (c). 1.88 mg/mL Na2HP04 x 7 H20; (d). 1.72 mg/mL m-cresol; (e). 1.5 mg/mL phenol; (f). 17.88 mg/mL glycerol; (g). sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4; and (h). water.
In one aspect, the pharmaceutical formulation of the invention consists of: (a). 3.5 mg/mL insulin aspart; (b). 0.0196 mg/mL Zn(ll); (c). 1.88 mg/mL Na2HP04 x 7 H20; (d). 1.72 mg/mL m-cresol; (e). 1.5 mg/mL phenol; (f). 17.88 mg/mL glycerol; (g). from 0.1 mg/mL to 0.5 mg/mL protamine sulfate; (h). sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4; and (i). water.
In one aspect, the invention provides a process for preparing the pharmaceutical formulation of the invention, wherein the components are mixed together in the form of a solution or suspension, the pH is adjusted to reach the desired pH, and water is added to reach the final volume. In one aspect, the invention provides a kit comprising one or more separate packages of (a), the pharmaceutical formulation of the invention; and (b). a medical device.
In one aspect, the invention provides as kit comprising one or more separate packages of (a), the pharmaceutical formulation if the invention; and (b). at least one further active pharmaceutical ingredient; (c). and optionally a medical device. In one aspect, the kit of the invention comprises a further active pharmaceutical ingredient which is an antidiabetic agent. In one aspect, the kit of the invention comprises a further active pharmaceutical ingredient which is an antidiabetic agent selected from the group consisting of: (a), a GLP-1 receptor agonist;
(b). a dual GLP-1 receptor/glucagon receptor agonist; (c). human FGF-21 ; (d). a FGF-21 analogue; (e). a FGF-21 derivative; (f). insulin; (g). human insulin; (h). an analogue of insulin; and (i). a derivative of insulin.
In one aspect, the kit of the invention comprises a further active pharmaceutical ingredient which is an analogue or derivative of insulin selected from the group consisting of fast acting insulin and long acting insulin. In one aspect, the kit of the invention comprises an analogue or derivative of insulin selected from the group consisting of fast acting insulin and long acting insulin, wherein the fast acting insulin is selected from the group consisting of insulin aspart, insulin lispro and insulin glulisine and wherein the long acting insulin is selected from the group consisting of insulin glargin, insulin detemir and insulin degludec.
In one aspect, the invention provides a pharmaceutical formulation or a kit for use in the treatment of diabetes mellitus.
In one aspect, the invention provides a pharmaceutical formulation or a kit for use in the treatment of hyperglycemia.
In one aspect, the invention provides a pharmaceutical formulation or a kit for use in lowering blood glucose level. In one aspect, the invention provides a method of treating diabetes mellitus in a subject in need thereof comprising administering the pharmaceutical formulation of the invention.
In one aspect, the invention provides a method of treating hyperglycemia in a subject in need thereof comprising administering the pharmaceutical formulation of the invention. In one aspect, the invention provides a method of lowering blood glucose levels in a subject in need thereof comprising administering the pharmaceutical formulation of the invention.
In one aspect, the invention provides a medical device for administering the pharmaceutical formulation of the invention to an animal and/or human.
The present invention is illustrated by the following Examples. However, it should be understood that the present invention is not limited to the specific details of these examples. EXAMPLES
Example 1
Manufacturing process (a) Zinc Oxide Solution
Zinc oxide Solution (containing Zn(ll) ) was prepared by suspending 0.8539 g zinc oxide in 500 mL water for injection and dissolving by adding 1 N hydrochloric acid. The solution is filled up with water for injection to final volume of 1000 mL. (b) Solution A
The final composition of Solution A is given in Table 1
Table 1 : Composition of Solution A
Figure imgf000027_0001
': 21.04 mg glycerol 85% corresponds to 17.88 mg glycerol.
Solution A was prepared as described in the following:
1. It was started with approximately 1000 g water for injection.
2. 18.8 g di-Natriumhydrogen-phosphat * 7 H20, 210.4 g Glycerol 85%, 15.0 g phenol and 17.2 g m-cresol were dissolved while stirring constantly.
3. Solution was filled up to approximately 1800 g with water for injection.
4. Solution was stirred for approximately 15 min using a magnetic stirrer.
5. pH was checked (pH should be 8.65). If pH value is not 8.65, the pH was adjusted to said range using hydrochloric acid 0.03 N or sodium hydroxide solution 1 N.
6. Solution was filled up to 2015 g (=2000 mL) with water for injection.
(c) Final Solution
The final composition of Final Solution is given in Table 2:
Table 2: Composition of Final Solution
Figure imgf000028_0001
( ': 21.04 mg glycerol 85% corresponds to 17.88 mg glycerol.
Final Solution was prepared as described in the following:
1. It was started with 300 mL water for injection.
2. 7.0 g insulin aspart was added to the 300 mL water for injection while stirring constantly (a suspension of insulin aspart in water for injection is formed).
3. pH value was checked.
4. pH value was changed to approximately 3.1 to 3.2 by adding hydrochloric acid 0.03 N or sodium hydroxide solution 0.02 N to dissolve the insulin aspart.
5. Solution was stirred for approximately 15 min using a magnetic stirrer.
6. 57 mL zinc oxide solution was added to the solution while stirring constantly.
7. Solution was filled up to 600 g with water for injection.
8. 400 mL Solution A was added slowly and carefully while stirring constantly.
9. pH was adjusted to 7.4 (range 7.2 to 7.6) using hydrochloric acid 0.03 N or sodium
hydroxide solution 0.02 N. 10. Solution was filled up to 2010 g with water for injection (corresponds to 100 % of the Final Solution).
Due to the addition of NaOH and HCI (for pH-adjustments and dissolving zinc oxide, see above), 0.17 mg/mL NaCI can be formed in the final solution
Quality control: Final solution was a clear and uncoloured solution, showed a pH value of 7.4 (plus/minus 0.2; at 20-25°C). The Final Solution was applied to sterile filtration using 'Sartopore Minisart high flow' filter (filter material: polyethersulfone; pore size: 0.2 μηι; supplier: Sartorius).
The Final Solution after sterile filtration was a clear and uncoloured solution and showed an osmolarity of 260 mOsmol/kg (plus/minus 30).
The Final Solution after sterile filtration was filled into appropriate vials (volume: 5 and 10 mL; 13 mm; clear glas; glas type 1 ).
The vials -containing the Final Solution after sterile filtration- were stored between +2°C and + 8°C and protected from light.
Example 2
Control of the formulation
(a) Analytical procedures
Tests are carried out using compendial analytical test methods, where applicable. The quality control concept has been established taking into account the cGMP requirements as well as the current status of the ICH process.
The non-compendial and chromatographic analytical procedures used to control the formulation are summarized in the following:
Description
Visually examine a number of containers for conformance to the acceptance criteria. Identification (HPLC)
The identity of the active ingredient is ensured by comparing the retention time of the drug formulation sample with the retention time of the reference standard using a reversed phase HPLC method. The method is also used for the determination of assay of the active ingredient, for the determination of the related compounds and impurities, and for quantifying the preservatives m- cresol and phenol.
Assay (HPLC)
The test is carried out by reverse phase liquid chromatography (HPLC). The method is also used for the identification, the determination of assay of the active ingredient, for the determination of the related compounds and impurities, and for quantifying the preservatives m-cresol and phenol. Column: Lichrosorb RP18, particle size 5 μηι, pore size 100 A (250 mm χ 4.0 mm), thermostated at +35 °C. Autosampler: Thermostated at <+8 °C. Mobile phase A: Sodium sulfate solved in water, 14 g/mL, adjusted with phosphoric acid and sodium hydroxide to a pH of 3.4. Mobile phase B: Water/acetonitrile (50:50 v/v). Gradient is shown in Table 3.
Table 3: HPLC gradient
Figure imgf000030_0001
Flow rate: 1.0 mL/min. Injection volume: 10 μί. Detection: 214 nm (for the active ingredient) and 260 nm (for m-cresol and phenol). Typical run time: 60 min.
Assay of the active ingredient, m-cresol and phenol are calculated by external standardization.
Impurities are calculated using the peak area percent method.
Test solution: The formulation is used without any dilution or further treatment. Related compounds and impurities (HPLC)
The same chromatographic conditions as for "Assay (HPLC)" are used for the determination of related compounds and impurities. Related compounds and Impurities are calculated using the peak area percent method.
High molecular weight proteins (HMWPs)
The high molecular weight proteins are determined using high pressure size exclusion
chromatography (HPSEC). Column: Waters Insulin HMWP, particle size 5-10 μηι, pore size 12 - 12.5 nm (300 mm χ 7.8 mm), thermostated at room temperature. Autosampler: thermostated at <+8°C. Mobile phase: 650 mL of arginine solution (1 g/L) is mixed with 200 mL of acetonitrile and 150 mL of glacial acetic acid. Isocratic elution Flow rate: 1.0 mL/min. Injection volume: 100 μί. Detection: 276 nm. Typical run time: 35 min.
HMWPs are calculated using the peak area percent method. Test solution: The formulation is used without any dilution or further treatment.
Antimicrobial preservative assay
The same chromatographic conditions as for "Assay (HPLC)" are used for the determination of assay of m-Cresol and of phenol m-cresol and phenol are calculated by external standardization.
(b) Validation of analytical procedures
The HPLC analytical procedure for the formulation for the determination of identification, assay, and related compounds and impurities was validated to demonstrate specificity, linearity, limit of detection and limit of quantification, accuracy, precision and range.
(c) Justification of the acceptance criteria
Tests and acceptance criteria, as previously presented, were selected based on ICH Q6B and on published monographs, analytical results obtained, precision of procedures used, Pharmacopoeial and/or regulatory guidelines, and are in agreement with the standard limits at this stage of development. Example 3
Stability of the formulation
(a) Stability of the formulation
Stability studies for the formulation were initiated according to the stability protocol summary described in the following table. The composition and manufacturing method of the stability batches are representative of the material. The stability profile was assessed for storage under long term, accelerated, and stress testing conditions according to ICH guidelines. Samples were packed and stored in glass vials with flanged cap with inserted disc and flip-off lid. The stability data obtained using this packaging material are representative for the preliminary shelf life and storage direction for both packaging configurations (10 ml. glass vials and 3 ml. cartridges).
1 month stability data are available from ongoing stability studies of the formulation.
Table 4: Storage Conditions
Figure imgf000032_0001
The following tests were performed during stability testing: appearance, assay, related impurities, high molecular weight proteins, pH, particulate matter (visible and subvisible particles), assay of antimicrobial preservatives (m-cresol and phenol), content of Zn(ll). The investigations on physical and chemical properties after 1 month of storage at long term storage condition of +5°C confirm the stability of the formulation when stored at the recommended storage condition. Only very slight changes of the related impurities could be observed.
When stored at accelerated conditions (1 months at +25°C/60%RH) the related impurities and high molecular weight proteins increased, however stayed well within the current acceptance limit. When stored at accelerated conditions (1 month at +40°C/75%RH) one of the related impurities increased above the acceptance criterion. The content of the active ingredient decreased below the acceptance criteria. The content of the microbial preservatives, m-cresol and phenol, remained basically unchanged under accelerated conditions.
When stored exposed to light (sun test according to ICH guidelines for 1 day and indoor light for 14 days) the related impurities and high molecular weight proteins increased above the acceptance criteria. The content of the active ingredient, m-cresol and phenol, remained basically unchanged after photostability testing.
Due to the present results of the stability studies of the formulation, the chemical and physical stability of the formulation can be confirmed.
Tables 5 and 6 show the long term stability results, wherein batch no. "_0021 " is referring to a formulation according to the present invention.
The stability of the formulation as presently claimed shows an excellent chemical and physical stability which qualifies said aqueous pharmaceutical formulation as medicinal product having a defined shelf life.
Table 5: Stability data of batch _0021 stored for 1 month at long term storage condition +5°C, at accelerated condition +25°C, and at stress condition +40°C
Figure imgf000034_0001
Table 6: Photostability Suntest batch
Storage condition: Suntest per ICH guideline and indoor light
Test item Acceptance Time
criteria for clinical Sun test (ICH) Indoor light
trials Initial value Dark 1 day Dark control 14 days control
Appearance of solution
(visual)
- Clarity Monitoring < I (water < I (water < I (water < I (water clear) < I (water clear) clear) clear) clear)
- Color Monitoring B9 B9 B5 B9 B7
Assay in insulin aspart 90.0 insulin aspart 107.7 insulin 108.0 insulin 91.4 insulin 102.9 insulin 96.0 insulin aspart units (HPLC) units/mL to 110.0 insulin aspart units/mL aspart units/mL aspart aspart units/mL units/mL aspart units/mL (3.77 mg/mL) (3.78 mg/mL) units/mL (3.60 mg/mL) (3.36 mg/mL)
(3.20 mg/mL)
Related
Compounds(HPLC)
- B28isoAsp insulin <2.5% 0.19% 0.29% 0.28% 0.45% 0.42% aspart
- Total of A21Asp insulin <5.0% 1.30% 1.26% 1.58% 1.57% 1.62% aspart, B3Asp insulin
aspart and B3isoAsp
insulin aspart
- Any other unspecified, <2.0% 0.36% 0.28% 1.09% 0.51% 0.66% unidentified
impurity
- Total of other impurities <3.5% 0.57% 0.84% 14.9% 0.86% 5.33%
High molecular weight <1.5% 0.21% 0.23% 6.57% 0.29% 4.07% proteins (HPSEC)
pH Between 7.0 to 7.8 7.42 7.44 7.41 7.41 7.40
Particulate matter (visible Practically free from complies Complies Complies Complies Complies particles) visible particles
Particulate matter Number of particles per
(subvisible particles) container 5 2 1 1 1
>10 μπι: 0 0 0 0 1
<6000
>25 μπι:
<600
Assay m-cresol 1.55 mg/mL to 1.89 1.76 mg/mL 1.75 mg/mL 1.72 mg/mL 1.75 mg/mL 1.74 mg/mL mg/mL (102.3%) (101.7%) (100.0%) (101.7%) (101.2%) (90.0% to 110.0% of
label claim)
Assay phenol 1.35 mg/mL to 1.65 1.49 mg/mL 1.50 mg/mL 1.48 mg/mL 1.48 mg/mL 1.48 mg/mL mg/mL (99.3%) (100.0%) (98.7%) (98.7%) (98.7%) (90.0% to 110.0% of
label claim)
Zinc (Zn (II) ) (AAS) <40 μg per 100 units 18.9 μg per 18.9 μg per 22.8 μg per 19.7 μg per 100 21.0 μg per 100 insulin aspart 100 units 100 units 100 units units insulin units insulin aspart insulin aspart insulin aspart insulin aspart aspart (20.2 μg/mL) (20.4 μg/mL) (20.4 μg/mL) (20.8 μg/mL) (20.3 μg/mL)

Claims

Claims
1. A pharmaceutical formulation comprising
(a) , insulin aspart; and
(b) . Zn(ll); and
(c) . optionally protamine;
wherein the pharmaceutical formulation contains less than 0.17 mg/mL sodium chloride.
The pharmaceutical formulation according to claim 1 , wherein the pharmaceutical formulation is an aqueous pharmaceutical formulation.
The pharmaceutical formulation according to claim 1 or 2, wherein the pharmaceutical formulation has a pH value in the range from 6.0 to 9.0.
The pharmaceutical formulation according to claim 3, wherein
the pharmaceutical formulation has a pH value in the range from 7.0 to 7.8.
The pharmaceutical formulation as claimed in one or more of claims 1 to 2, wherein insulin aspart is present in a concentration from 10 U/mL to 1000 U/mL.
The pharmaceutical formulation as claimed in one or more of claims 1 to 3, wherein Zn(ll) is present in a concentration from 0.0100 to 0.0600 mg / 100 U of insulin aspart.
The pharmaceutical formulation as claimed in one or more of claims 1 to 4, wherein the pharmaceutical formulation is essentially free of sodium chloride.
The pharmaceutical formulation as claimed in one or more of claims 1 to 5, wherein protamine is present in a concentration from 0.1 to 0.5 mg/mL.
The pharmaceutical formulation as claimed in one or more of claims 1 to 6, wherein the pharmaceutical formulation further contains a buffering agent.
10. The pharmaceutical formulation as claimed in one or more of claims 1 to 7, wherein the pharmaceutical formulation comprises one or more further active pharmaceutical ingredients.
1 1. The pharmaceutical formulation according to claim 8, wherein the further active
pharmaceutical ingredient is an antidiabetic agent.
12. The pharmaceutical formulation according to claim 8 or 9, wherein the further active
pharmaceutical ingredient is an antidiabetic agent selected from the group consisting of: (a), a GLP-1 receptor agonist;
(b) . a dual GLP-1 receptor/glucagon receptor agonist;
(c) . human FGF-21 ;
(d) . a FGF-21 analogue;
(e) . a FGF-21 derivative;
(f). insulin;
(g) . human insulin;
(h) . an analogue of insulin; and
(i) . a derivative of insulin.
13. The pharmaceutical formulation as claimed in one or more of claims 1 to 10, wherein the pharmaceutical formulation further comprises a fast acting analogue and/or derivative of insulin or a long acting analogue and/or derivative of insulin.
14. The pharmaceutical formulation according to claim 1 1 , wherein the fast acting insulin is one or more insulin selected from the group consisting of insulin lispro and insulin glulisine and wherein the long acting insulin is one or more insulin selected from the group consisting of insulin detemir, insulin glargin and insulin degludec.
15. The pharmaceutical formulation as claimed in one or more of claims 1 to 5, wherein the pharmaceutical formulation consists of:
(a) . 3.5 mg/ml_ insulin aspart;
(b) . 0.0196 mg/ml_ Zn(ll) ;
(c) . 1.88 mg/mL Na2HP04 x 7 H20;
(d) . 1.72 mg/mL m-cresol;
(e). 1.5 mg/mL phenol; (f) . 17.88 mg/mL glycerol;
(g) . sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4; and
(h) . water.
16. The pharmaceutical formulation as claimed in one or more of claims 1 to 5, wherein the pharmaceutical formulation consists of:
(a) . 3.5 mg/mL insulin aspart;
(b) . 0.0196 mg/mL Zn(ll);
(c) . 1.88 mg/mL Na2HP04 x 7 H20;
(d) . 1.72 mg/mL m-cresol;
(e) . 1.5 mg/mL phenol;
(f) . 17.88 mg/mL glycerol;
(g) . from 0.1 mg/mL to 0.5 mg/mL protamine sulfate;
(h) . sodium hydroxide and/or hydrochloric acid to adjust the pH to 7.4; and
(i) . water.
17. A process for preparing the pharmaceutical formulation according to claim 1 , wherein the components are mixed together in the form of an solution or suspension, the pH is adjusted to reach the desired pH, and water is added to reach the final volume.
18. A kit comprising one or more separate packages of
(a) , the pharmaceutical formulation as claimed in one or more of claims 1 to 14; and
(b) . a medical device.
19. A kit comprising one or more separate packages of
(a) , the pharmaceutical formulation as claimed in one or more of claims 1 to 14; and
(b) . at least one further active pharmaceutical ingredient;
(c) . and optionally a medical device.
20. The kit according to one or more of claims 16-17, wherein the further active
pharmaceutical ingredient is an antidiabetic agent.
21 The kit as claimed in one or more of claims 17 to 18, wherein the further active
pharmaceutical ingredient is an antidiabetic agent selected from the group consisting of: (a) , a GLP-1 receptor agonist;
(b) . a dual GLP-1 receptor/glucagon receptor agonist;
(c) . human FGF-21 ;
(d) . a FGF-21 analogue;
(e) . a FGF-21 derivative;
(f) . insulin;
(g) . human insulin;
(h) . an analogue of insulin; and
(i) . a derivative of insulin.
22. The kit as claimed in one or more of claims 17 to 19, wherein the further active
pharmaceutical ingredient is an analogue or derivative of insulin selected from the group consisting of fast acting insulin and long acting insulin.
23. The kit according to claim 20, wherein the fast acting insulin is selected from the group consisting of insulin aspart, insulin lispro and insulin glulisine and wherein the long acting insulin is selected from the group consisting of insulin glargin, insulin detemir and insulin degludec.
24. The pharmaceutical formulation as claimed in one or more of claims 1 to 14 or the kit as claimed in one or more of claims 16 to 21 for use in the treatment of diabetes mellitus.
25. The pharmaceutical formulation as claimed in one or more of claims 1 to 14 or the kit as claimed in one or more of claims 16 to 21 for use in the treatment of hyperglycemia.
26. The pharmaceutical formulation as claimed in one or more of claims 1 to 14 or the kit as claimed in one or more of claims 16 to 21 for use in lowering blood glucose level.
27. A method of treating diabetes mellitus in a subject in need thereof comprising
administering the pharmaceutical formulation of any one of claims 1 to 14.
28. A method of treating hyperglycemia in a subject in need thereof comprising administering the pharmaceutical formulation of any one of claims 1 to 14.
29. A method of lowering blood glucose levels in a subject in need thereof comprising
administering the pharmaceutical formulation of any one of claims 1 to 14.
30. Medical device for administering the pharmaceutical formulation as claimed in one or more of claims 1 to 14 to an animal and/or human.
PCT/EP2015/050215 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart WO2015104310A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
BR112016015851A BR112016015851A2 (en) 2014-01-09 2015-01-08 STABILIZED PHARMACEUTICAL FORMULATIONS OF INSULIN ASPART
RU2016132340A RU2016132340A (en) 2014-01-09 2015-01-08 STABILIZED PHARMACEUTICAL COMPOSITIONS BASED ON INSULIN ASPART
AU2015205620A AU2015205620A1 (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart
SG11201604706TA SG11201604706TA (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart
JP2016544426A JP6735674B2 (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulation of insulin aspart
CN201580003940.9A CN105899190B (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart
EP15700071.2A EP3091964A1 (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart
CN202210571205.XA CN114939156A (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart
KR1020167021373A KR20160101195A (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart
CA2932873A CA2932873A1 (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart
MX2016008977A MX2016008977A (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart.

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201461925472P 2014-01-09 2014-01-09
EP14305023 2014-01-09
EP14305023.5 2014-01-09
US61/925,472 2014-01-09

Publications (1)

Publication Number Publication Date
WO2015104310A1 true WO2015104310A1 (en) 2015-07-16

Family

ID=49949600

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/050215 WO2015104310A1 (en) 2014-01-09 2015-01-08 Stabilized pharmaceutical formulations of insulin aspart

Country Status (12)

Country Link
US (3) US9895423B2 (en)
EP (1) EP3091964A1 (en)
JP (2) JP6735674B2 (en)
KR (1) KR20160101195A (en)
CN (2) CN105899190B (en)
AU (1) AU2015205620A1 (en)
BR (1) BR112016015851A2 (en)
CA (1) CA2932873A1 (en)
MX (1) MX2016008977A (en)
RU (1) RU2016132340A (en)
SG (1) SG11201604706TA (en)
WO (1) WO2015104310A1 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI641381B (en) 2013-02-04 2018-11-21 法商賽諾菲公司 Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
TW201605489A (en) * 2013-10-25 2016-02-16 賽諾菲公司 Stable formulation of INSULIN GLULISINE
WO2015104314A1 (en) 2014-01-09 2015-07-16 Sanofi Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
SG11201604708VA (en) * 2014-01-09 2016-07-28 Sanofi Sa Stabilized glycerol free pharmaceutical formulations of insulin analogues and/or insulin derivatives
JP2020143105A (en) * 2014-01-09 2020-09-10 サノフイSanofi Stabilized pharmaceutical formulations of insulin aspart
KR20160101195A (en) * 2014-01-09 2016-08-24 사노피 Stabilized pharmaceutical formulations of insulin aspart
GB201607918D0 (en) * 2016-05-06 2016-06-22 Arecor Ltd Novel formulations
JP2019529506A (en) * 2016-09-29 2019-10-17 アレコル リミテッド New formulation
SG11202008971VA (en) * 2018-03-16 2020-10-29 Anji Pharmaceuticals Inc Compositions and methods for treating severe constipation
CN114306577B (en) * 2020-10-10 2024-04-09 南京汉欣医药科技有限公司 Preparation method of insulin aspart 30 suspension
TW202241492A (en) * 2020-12-16 2022-11-01 丹麥商西蘭製藥公司 Pharmaceutical composition of glp-1/glp-2 dual agonists
CN113896784B (en) * 2021-10-18 2024-04-16 合肥天麦生物科技发展有限公司 Preparation method of insulin crystal and product thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995000550A1 (en) * 1993-06-21 1995-01-05 Novo Nordisk A/S Aspb28 insulin crystals
WO1997048413A1 (en) * 1996-06-20 1997-12-24 Novo Nordisk A/S Insulin preparations containing carbohydrates
WO1999021578A1 (en) * 1997-10-24 1999-05-06 Eli Lilly And Company Insoluble insulin compositions
WO2006029634A2 (en) * 2004-09-17 2006-03-23 Novo Nordisk A/S Pharmaceutical compositions containing insulin and insulinotropic peptide
WO2011012719A1 (en) * 2009-07-31 2011-02-03 Ascendis Pharma As Long acting insulin composition
WO2012080320A1 (en) * 2010-12-14 2012-06-21 Novo Nordisk A/S Fast-acting insulin in combination with long-acting insulin

Family Cites Families (151)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB835638A (en) 1956-12-01 1960-05-25 Novo Terapeutisk Labor As Insulin crystal suspensions having a protracted effect
GB840870A (en) 1957-08-03 1960-07-13 Novo Terapeutisk Labor As Improvements in or relating to insulin preparations
JPS6033474B2 (en) 1978-05-11 1985-08-02 藤沢薬品工業株式会社 Novel hyaluronidase BMP-8231 and its production method
PH25772A (en) 1985-08-30 1991-10-18 Novo Industri As Insulin analogues, process for their preparation
CA1275922C (en) * 1985-11-28 1990-11-06 Harunobu Amagase Treatment of cancer
US5614492A (en) 1986-05-05 1997-03-25 The General Hospital Corporation Insulinotropic hormone GLP-1 (7-36) and uses thereof
DK257988D0 (en) 1988-05-11 1988-05-11 Novo Industri As NEW PEPTIDES
DE3837825A1 (en) 1988-11-08 1990-05-10 Hoechst Ag NEW INSULIN DERIVATIVES, THEIR USE AND A PHARMACEUTICAL PREPARATION CONTAINING THEM
HUT56857A (en) 1988-12-23 1991-10-28 Novo Nordisk As Human insulin analogues
PT93057B (en) 1989-02-09 1995-12-29 Lilly Co Eli PROCESS FOR THE PREPARATION OF INSULIN ANALOGS
ATE133087T1 (en) 1989-05-04 1996-02-15 Southern Res Inst ENCAPSULATION PROCESS
US5545618A (en) 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
DK155690D0 (en) 1990-06-28 1990-06-28 Novo Nordisk As NEW PEPTIDES
US5272135A (en) 1991-03-01 1993-12-21 Chiron Ophthalmics, Inc. Method for the stabilization of methionine-containing polypeptides
DK36392D0 (en) 1992-03-19 1992-03-19 Novo Nordisk As USE OF CHEMICAL COMPOUND
DK39892D0 (en) 1992-03-25 1992-03-25 Bernard Thorens PEPTIDE
US5846747A (en) 1992-03-25 1998-12-08 Novo Nordisk A/S Method for detecting glucagon-like peptide-1 antagonists and agonists
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
PT729353E (en) 1993-11-19 2002-07-31 Alkermes Inc PREPARATION OF BIODEGRADABLE MICROPARTICLES CONTAINING A BIOLOGICALLY ACTIVE AGENT
US5705483A (en) 1993-12-09 1998-01-06 Eli Lilly And Company Glucagon-like insulinotropic peptides, compositions and methods
AU723694B2 (en) 1996-06-05 2000-08-31 Roche Diagnostics Gmbh Exendin analogues, processes for their preparation and medicaments containing them
DE19637230A1 (en) 1996-09-13 1998-03-19 Boehringer Mannheim Gmbh Truncated versions of exendin peptide(s) for treating diabetes
US5948751A (en) * 1996-06-20 1999-09-07 Novo Nordisk A/S X14-mannitol
ATE208208T1 (en) * 1996-06-20 2001-11-15 Novo Nordisk As INSULIN PREPARATIONS CONTAINING HALOGENIDES
US6110703A (en) 1996-07-05 2000-08-29 Novo Nordisk A/S Method for the production of polypeptides
DE69739172D1 (en) 1996-08-08 2009-01-29 Amylin Pharmaceuticals Inc REGULATION OF GASTROINTESTINAL MOBILITY
US6006753A (en) 1996-08-30 1999-12-28 Eli Lilly And Company Use of GLP-1 or analogs to abolish catabolic changes after surgery
US6384016B1 (en) 1998-03-13 2002-05-07 Novo Nordisk A/S Stabilized aqueous peptide solutions
US6268343B1 (en) 1996-08-30 2001-07-31 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6277819B1 (en) 1996-08-30 2001-08-21 Eli Lilly And Company Use of GLP-1 or analogs in treatment of myocardial infarction
UA65549C2 (en) 1996-11-05 2004-04-15 Елі Ліллі Енд Компані Use of glucagon-like peptides such as glp-1, glp-1 analog, or glp-1 derivative in methods and compositions for reducing body weight
WO1998030231A1 (en) 1997-01-07 1998-07-16 Amylin Pharmaceuticals, Inc. Use of exendins and agonists thereof for the reduction of food intake
WO1998035033A1 (en) 1997-02-05 1998-08-13 1149336 Ontario Inc. Polynucleotides encoding proexendin, and methods and uses thereof
US5846937A (en) 1997-03-03 1998-12-08 1149336 Ontario Inc. Method of using exendin and GLP-1 to affect the central nervous system
PE79099A1 (en) * 1997-06-13 1999-08-24 Lilly Co Eli STABLE INSULIN FORMULATIONS
CA2292730C (en) 1997-06-13 2008-09-16 Genentech, Inc. Stabilized antibody formulation
EP1019077B2 (en) 1997-08-08 2010-12-22 Amylin Pharmaceuticals, Inc. Novel exendin agonist compounds
US6444641B1 (en) 1997-10-24 2002-09-03 Eli Lilly Company Fatty acid-acylated insulin analogs
AU756836B2 (en) 1997-11-14 2003-01-23 Amylin Pharmaceuticals, Inc. Novel exendin agonist compounds
DE69838916T2 (en) 1997-11-14 2008-12-18 Amylin Pharmaceuticals, Inc., San Diego NOVEL EXENDIN AGONISTS
JP2001525371A (en) 1997-12-05 2001-12-11 イーライ・リリー・アンド・カンパニー GLP-1 preparation
JP4677095B2 (en) 1998-02-13 2011-04-27 アミリン・ファーマシューティカルズ,インコーポレイテッド Inotropic and diuretic effects of exendin and GLP-1
EP1056775B1 (en) 1998-02-27 2010-04-28 Novo Nordisk A/S Glp-1 derivatives of glp-1 and exendin with protracted profile of action
EP1950224A3 (en) 1998-03-09 2008-12-17 Zealand Pharma A/S Pharmacologically active peptide conjugates having a reduced tendency towards enzymatic hydrolysis
ES2320715T3 (en) 1998-06-05 2009-05-27 Nutrinia Limited ENRICHED CHILD MILK WITH INSULIN SUPPLEMENT.
EP1119625B1 (en) 1998-10-07 2005-06-29 Medical College Of Georgia Research Institute, Inc. Glucose-dependent insulinotropic peptide for use as an osteotropic hormone
US6284725B1 (en) 1998-10-08 2001-09-04 Bionebraska, Inc. Metabolic intervention with GLP-1 to improve the function of ischemic and reperfused tissue
US6211144B1 (en) 1998-10-16 2001-04-03 Novo Nordisk A/S Stable concentrated insulin preparations for pulmonary delivery
US6489292B1 (en) 1998-11-18 2002-12-03 Novo Nordisk A/S Stable aqueous insulin preparations without phenol and cresol
JP2000247903A (en) 1999-03-01 2000-09-12 Chugai Pharmaceut Co Ltd Long-term stabilized pharmaceutical preparation
JP2007204498A (en) 1999-03-01 2007-08-16 Chugai Pharmaceut Co Ltd Long-term stabilized formulations
US6271241B1 (en) 1999-04-02 2001-08-07 Neurogen Corporation Cycloalkyl and aryl fused aminoalkyl-imidazole derivatives: modulators and GLP-1 receptors
CN1372570A (en) 1999-04-30 2002-10-02 安米林药品公司 Modified exendin and exendin agonists
PT1180121E (en) 1999-05-17 2004-03-31 Conjuchem Inc LONG-TERM INSULINOTROPIC PEPTIDES
US6344180B1 (en) 1999-06-15 2002-02-05 Bionebraska, Inc. GLP-1 as a diagnostic test to determine β-cell function and the presence of the condition of IGT and type II diabetes
US6528486B1 (en) 1999-07-12 2003-03-04 Zealand Pharma A/S Peptide agonists of GLP-1 activity
EP1076066A1 (en) 1999-07-12 2001-02-14 Zealand Pharmaceuticals A/S Peptides for lowering blood glucose levels
DE19947456A1 (en) 1999-10-02 2001-04-05 Aventis Pharma Gmbh New synthetic derivatives of the C-peptide of proinsulin, useful in the preparation of human insulin or insulin analogs in high yield
CN1245216C (en) 1999-10-04 2006-03-15 希龙公司 Stabilized liquid polypeptide-confg. pharmaceutical compositions
US20010012829A1 (en) 2000-01-11 2001-08-09 Keith Anderson Transepithelial delivery GLP-1 derivatives
WO2001051071A2 (en) 2000-01-11 2001-07-19 Novo Nordisk A/S Transepithelial delivery of glp-1 derivatives
US6395767B2 (en) 2000-03-10 2002-05-28 Bristol-Myers Squibb Company Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl peptidase IV and method
US6852694B2 (en) 2001-02-21 2005-02-08 Medtronic Minimed, Inc. Stabilized insulin formulations
DE10114178A1 (en) 2001-03-23 2002-10-10 Aventis Pharma Gmbh Zinc-free and low-zinc insulin preparations with improved stability
DE60233722D1 (en) 2001-04-02 2009-10-29 Novo Nordisk As INSULIN PREPARATIONS AND METHOD FOR THE PRODUCTION THEREOF
WO2003002021A2 (en) 2001-06-29 2003-01-09 The Regents Of The University Of California Biodegradable/bioactive nucleus pulposus implant and method for treating degenerated intervertebral discs
JP2005508895A (en) 2001-08-28 2005-04-07 イーライ・リリー・アンド・カンパニー Premix of GLP-1 and basal insulin
CA2468100A1 (en) 2001-12-20 2003-07-03 Eli Lilly And Company Insulin molecule having protracted time action
AU2003203146A1 (en) 2002-02-07 2003-09-02 Novo Nordisk A/S Use of glp-1 compound for treatment of critically ill patients
EP1506003A1 (en) 2002-05-07 2005-02-16 Novo Nordisk A/S Soluble formulations comprising insulin aspart and insulin detemir
WO2003094956A1 (en) 2002-05-07 2003-11-20 Novo Nordisk A/S Soluble formulations comprising monomeric insulin and acylated insulin
DE10227232A1 (en) 2002-06-18 2004-01-15 Aventis Pharma Deutschland Gmbh Sour insulin preparations with improved stability
EP1525219B1 (en) 2002-07-04 2009-05-27 Zealand Pharma A/S Glp-1 and methods for treating diabetes
BR0314996A (en) 2002-10-02 2005-08-09 Zealand Pharma As Composition, pharmaceutically acceptable composition, method for producing the composition, methods for stabilizing exendin-4 (1-39) or a variant, derivative or analogue thereof against degradation, before, during or after intended use, to treat diseases, to treat disease states associated with elevated blood glucose levels, to regulate blood glucose levels, to regulate gastric emptying, to stimulate the release of insulin in a mammal to reduce blood glucose level in a mammal, to reduce the level of plasma lipids in a mammal, to reduce mortality and morbidity after myocardial infarction in a mammal, to stimulate insulin release in a mammal, and to produce a stabilized exendin (1-39), and stabilized exendin (1-39)
MXPA05009565A (en) 2003-03-11 2005-12-02 Novo Nordisk As Pharmaceutical preparations comprising acid-stabilised insulin.
DE10325567B4 (en) 2003-06-05 2008-03-13 Mavig Gmbh Radiation protection arrangement with separable enclosure
EP1663295A2 (en) 2003-09-01 2006-06-07 Novo Nordisk A/S Stable formulations of peptides
JP2007537981A (en) 2003-09-19 2007-12-27 ノボ ノルディスク アクティーゼルスカブ Novel plasma protein affinity tag
ATE525083T1 (en) 2003-11-13 2011-10-15 Novo Nordisk As PHARMACEUTICAL COMPOSITION COMPRISING AN INSULINOTROPIC GLP-1(7-37) ANALOG, ASP(B28) INSULIN, AND A SURFACE-ACTIVE COMPOUND
US20050201978A1 (en) 2003-11-17 2005-09-15 Lipton James S. Tumor and infectious disease therapeutic compositions
US20080090753A1 (en) 2004-03-12 2008-04-17 Biodel, Inc. Rapid Acting Injectable Insulin Compositions
EP1758597B1 (en) 2004-05-20 2012-09-12 Diamedica Inc. Use of drug combinations for treating insulin resistance
US20060073213A1 (en) 2004-09-15 2006-04-06 Hotamisligil Gokhan S Reducing ER stress in the treatment of obesity and diabetes
CN106137952B (en) 2004-11-12 2020-11-17 诺和诺德公司 Stable formulations of insulinotropic peptides
JP5248113B2 (en) 2004-11-12 2013-07-31 ノヴォ ノルディスク アー/エス Peptide stable formulation
ES2391776T3 (en) * 2004-11-22 2012-11-29 Novo Nordisk A/S Soluble, stable formulations containing insulin with a protamine salt
DE102004058306A1 (en) 2004-12-01 2006-07-27 Sanofi-Aventis Deutschland Gmbh Process for the preparation of carboxy-terminally amidated peptides
EP1849024A4 (en) 2005-02-01 2015-08-26 Canberra Ind Inc Maximum entropy signal detection method
EP1909824B1 (en) 2005-04-08 2011-05-18 Amylin Pharmaceuticals, Inc. Pharmaceutical formulations comprising incretin peptide and aprotic polar solvent
US8097584B2 (en) 2005-05-25 2012-01-17 Novo Nordisk A/S Stabilized formulations of insulin that comprise ethylenediamine
WO2006131730A1 (en) 2005-06-06 2006-12-14 Camurus Ab Glp-1 analogue formulations
DE102005046113A1 (en) 2005-09-27 2007-03-29 Sanofi-Aventis Deutschland Gmbh Preparation of C-amidated peptides, useful as pharmaceuticals, by reaction between precursor peptides in presence of enzyme with activity of trypsin, also new reaction products
KR101105871B1 (en) 2005-09-27 2012-01-16 주식회사 엘지생명과학 hFSF Aqueous Formulation
US20090264732A1 (en) 2005-10-11 2009-10-22 Huntington Medical Research Institutes Imaging agents and methods of use thereof
US8343914B2 (en) 2006-01-06 2013-01-01 Case Western Reserve University Fibrillation resistant proteins
US20090324701A1 (en) 2006-01-20 2009-12-31 Diamedica, Inc. Compositions containing (s)-bethanechol and their use in the treatment of insulin resistance, type 2 diabetes, glucose intolerance and related disorders
US20070191271A1 (en) 2006-02-10 2007-08-16 Dow Pharmaceutical Sciences Method for stabilizing polypeptides lacking methionine
EP1986674A4 (en) 2006-02-13 2009-11-11 Nektar Therapeutics Methionine-containing protein or peptide compositions and methods of making and using
WO2007104786A1 (en) 2006-03-15 2007-09-20 Novo Nordisk A/S Mixtures of amylin and insulin
JP2009532422A (en) 2006-04-03 2009-09-10 ノボ・ノルデイスク・エー/エス GLP-1 peptide agonist
NZ571862A (en) 2006-04-13 2011-10-28 Ipsen Pharma Sas Pharmaceutical composition comprising HGLP-1, a zinc divalent metal ion and a solvent
DE102006031962A1 (en) 2006-07-11 2008-01-17 Sanofi-Aventis Deutschland Gmbh Amidated insulin glargine
US20090318353A1 (en) 2006-08-25 2009-12-24 Novo Nordisk A/S Acylated Exendin-4 Compounds
BRPI0717098B8 (en) 2006-09-22 2021-05-25 Novo Nordisk As protease-resistant human insulin analogues, pharmaceutical composition and process for their preparation
WO2008124522A2 (en) 2007-04-04 2008-10-16 Biodel, Inc. Amylin formulations
EP2157967B1 (en) 2007-04-23 2013-01-16 Intarcia Therapeutics, Inc Suspension formulations of insulinotropic peptides and uses thereof
CN101366692A (en) 2007-08-15 2009-02-18 江苏豪森药业股份有限公司 Stable Exenatide formulation
GB0717399D0 (en) 2007-09-07 2007-10-17 Uutech Ltd Use of GLP-1 analogues for the treatment of disorders associated with dysfunctional synaptic transmission
US8664369B2 (en) 2007-11-01 2014-03-04 Merck Serono S.A. LH liquid formulations
WO2009060071A1 (en) * 2007-11-08 2009-05-14 Novo Nordisk A/S Insulin derivative
ES2612736T3 (en) 2007-11-16 2017-05-18 Novo Nordisk A/S Stable pharmaceutical compositions comprising liraglutide and degludec
CN101444618B (en) 2007-11-26 2012-06-13 杭州九源基因工程有限公司 Pharmaceutical preparation containing exenatide
CN102026666B (en) 2007-12-11 2013-10-16 常山凯捷健生物药物研发(河北)有限公司 Formulation of insulinotropic peptide conjugates
PL2229407T3 (en) 2008-01-09 2017-06-30 Sanofi-Aventis Deutschland Gmbh Novel insulin derivatives having an extremely delayed time-action profile
CN101970476B (en) 2008-01-09 2014-08-27 塞诺菲-安万特德国有限公司 Novel insulin derivatives having an extremely delayed time-action profile
WO2009102467A2 (en) 2008-02-13 2009-08-20 Intarcia Therapeutics, Inc. Devices, formulations, and methods for delivery of multiple beneficial agents
TWI394580B (en) 2008-04-28 2013-05-01 Halozyme Inc Super fast-acting insulin compositions
KR100928942B1 (en) * 2008-05-26 2009-11-30 제이에스아이실리콘주식회사 Preparation method of linear and cyclic trisilaalkanes
US20100068210A1 (en) 2008-09-10 2010-03-18 Ji Junyan A Compositions and methods for the prevention of oxidative degradation of proteins
CN101670096B (en) 2008-09-11 2013-01-16 杭州九源基因工程有限公司 Medicinal preparation containing exenatide
JP5643762B2 (en) 2008-10-15 2014-12-17 インターシア セラピューティクス,インコーポレイティド High concentration drug particles, formulations, suspensions and their use
KR101820024B1 (en) 2008-10-17 2018-01-18 사노피-아벤티스 도이칠란트 게엠베하 Combination of an insulin and a GLP-1 agonist
JP2009091363A (en) 2008-11-21 2009-04-30 Asahi Kasei Pharma Kk Stabilized aqueous injectable solution of pth
SG10201403840VA (en) 2009-07-06 2014-10-30 Sanofi Aventis Deutschland Aqueous insulin preparations containing methionine
JP5675799B2 (en) 2009-07-06 2015-02-25 サノフィ−アベンティス・ドイチュラント・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング Slow-acting insulin preparation
WO2011003820A1 (en) 2009-07-06 2011-01-13 Sanofi-Aventis Deutschland Gmbh Heat- and vibration-stable insulin preparations
AR078161A1 (en) 2009-09-11 2011-10-19 Hoffmann La Roche VERY CONCENTRATED PHARMACEUTICAL FORMULATIONS OF AN ANTIBODY ANTI CD20. USE OF THE FORMULATION. TREATMENT METHOD
EP3345593B1 (en) 2009-11-13 2023-09-06 Sanofi-Aventis Deutschland GmbH Pharmaceutical composition comprising despro36exendin-4(1-39)-lys6-nh2 and methionine
US20110118180A1 (en) 2009-11-13 2011-05-19 Sanofi-Aventis Deutschland Gmbh Method of treatment of diabetes type 2 comprising add-on therapy to metformin
US20110118178A1 (en) 2009-11-13 2011-05-19 Sanofi-Aventis Deutschland Gmbh Method of treatment of diabetes type 2 comprising add-on therapy to insulin glargine and metformin
TR201809460T4 (en) 2009-11-13 2018-07-23 Sanofi Aventis Deutschland Pharmaceutical composition comprising a GLP-1-agonist, an insulin and methionine.
AU2011202239C1 (en) 2010-05-19 2017-03-16 Sanofi Long-acting formulations of insulins
EP2389945A1 (en) 2010-05-28 2011-11-30 Sanofi-Aventis Deutschland GmbH Pharmaceutical composition comprising AVE0010 and insulin glargine
ES2606554T3 (en) 2010-08-30 2017-03-24 Sanofi-Aventis Deutschland Gmbh Use of AVE0010 for the manufacture of a medication for the treatment of type 2 diabetes mellitus
JP6381914B2 (en) 2011-02-02 2018-08-29 サノフィ−アベンティス・ドイチュラント・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング Prevention of hypoglycemia in patients with type 2 diabetes
US20120277147A1 (en) 2011-03-29 2012-11-01 Sanofi-Aventis Deutschland Gmbh Prevention of hypoglycaemia in diabetes mellitus type 2 patients
US20130065828A1 (en) 2011-05-13 2013-03-14 Sanofi-Aventis Deutschland Gmbh Pharmaceutical combination for use in glycemic control in diabetes type 2 patients
US8735349B2 (en) 2011-05-13 2014-05-27 Sanofi-Aventis Deutschland Gmbh Method for improving glucose tolerance in a diabetes type 2 patient of younger than 50 years and having postprandial plasma glucose concentration of at least 14 mmol/L
US9821032B2 (en) 2011-05-13 2017-11-21 Sanofi-Aventis Deutschland Gmbh Pharmaceutical combination for improving glycemic control as add-on therapy to basal insulin
US20130040878A1 (en) 2011-05-13 2013-02-14 Sanofi-Aventis Deutschland Gmbh Pharmaceutical combination for use in the treatment of diabetes type 2 patients
TWI608847B (en) 2011-08-29 2017-12-21 賽諾菲阿凡提斯德意志有限公司 Pharmaceutical combination for use in glycemic control in diabetes type 2 patients
EP2763691A1 (en) 2011-10-04 2014-08-13 Sanofi-Aventis Deutschland GmbH Glp-1 agonist for use in the treatment of stenosis or/and obstruction in the biliary tract
PL2763690T3 (en) 2011-10-04 2016-04-29 Sanofi Aventis Deutschland Lixisenatide for use in the treatment of stenosis or/and obstruction in the pancreatic duct system
AU2012328388B2 (en) 2011-10-28 2017-06-15 Sanofi-Aventis Deutschland Gmbh Treatment protocol of diabetes type 2
US8901484B2 (en) 2012-04-27 2014-12-02 Sanofi-Aventis Deutschland Gmbh Quantification of impurities for release testing of peptide products
AR092862A1 (en) 2012-07-25 2015-05-06 Hanmi Pharm Ind Co Ltd LIQUID FORMULATION OF PROLONGED ACTION INSULIN AND AN INSULINOTROPIC PEPTIDE AND PREPARATION METHOD
TWI641381B (en) 2013-02-04 2018-11-21 法商賽諾菲公司 Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
GB201303771D0 (en) 2013-03-04 2013-04-17 Midatech Ltd Nanoparticles peptide compositions
WO2015104314A1 (en) 2014-01-09 2015-07-16 Sanofi Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
KR20160101195A (en) 2014-01-09 2016-08-24 사노피 Stabilized pharmaceutical formulations of insulin aspart
SG11201604708VA (en) 2014-01-09 2016-07-28 Sanofi Sa Stabilized glycerol free pharmaceutical formulations of insulin analogues and/or insulin derivatives

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995000550A1 (en) * 1993-06-21 1995-01-05 Novo Nordisk A/S Aspb28 insulin crystals
WO1997048413A1 (en) * 1996-06-20 1997-12-24 Novo Nordisk A/S Insulin preparations containing carbohydrates
WO1999021578A1 (en) * 1997-10-24 1999-05-06 Eli Lilly And Company Insoluble insulin compositions
WO2006029634A2 (en) * 2004-09-17 2006-03-23 Novo Nordisk A/S Pharmaceutical compositions containing insulin and insulinotropic peptide
WO2011012719A1 (en) * 2009-07-31 2011-02-03 Ascendis Pharma As Long acting insulin composition
WO2012080320A1 (en) * 2010-12-14 2012-06-21 Novo Nordisk A/S Fast-acting insulin in combination with long-acting insulin

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NOVO NORDISK INC.: "NovoLog Highlights of Prescribing Information", 12 October 2009 (2009-10-12), XP002724352, Retrieved from the Internet <URL:http://www.novonordisk-us.com/images/pdf/novolog_oct_12_2009.pdf> [retrieved on 20140514] *

Also Published As

Publication number Publication date
US20180125944A1 (en) 2018-05-10
CN114939156A (en) 2022-08-26
JP2017503797A (en) 2017-02-02
KR20160101195A (en) 2016-08-24
US20220023392A1 (en) 2022-01-27
JP2022180537A (en) 2022-12-06
CN105899190B (en) 2022-06-14
US20150190475A1 (en) 2015-07-09
MX2016008977A (en) 2016-10-04
AU2015205620A1 (en) 2016-07-14
RU2016132340A3 (en) 2018-09-18
CN105899190A (en) 2016-08-24
JP6735674B2 (en) 2020-08-05
EP3091964A1 (en) 2016-11-16
CA2932873A1 (en) 2015-07-16
BR112016015851A2 (en) 2017-08-08
RU2016132340A (en) 2018-02-14
US9895423B2 (en) 2018-02-20
SG11201604706TA (en) 2016-07-28

Similar Documents

Publication Publication Date Title
US20230049480A1 (en) Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
US10610595B2 (en) Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
US9895424B2 (en) Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
US9895423B2 (en) Stabilized pharmaceutical formulations of insulin aspart
JP2020128395A (en) Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
JP2020073534A (en) Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives

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: 15700071

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2932873

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2016544426

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/008977

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2015205620

Country of ref document: AU

Date of ref document: 20150108

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2015700071

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016015851

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 2015700071

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20167021373

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2016132340

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016015851

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160707