US20110301081A1 - Long-acting formulations of insulins - Google Patents

Long-acting formulations of insulins Download PDF

Info

Publication number
US20110301081A1
US20110301081A1 US13/110,568 US201113110568A US2011301081A1 US 20110301081 A1 US20110301081 A1 US 20110301081A1 US 201113110568 A US201113110568 A US 201113110568A US 2011301081 A1 US2011301081 A1 US 2011301081A1
Authority
US
United States
Prior art keywords
insulin
study
insulin glargine
subjects
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/110,568
Other languages
English (en)
Inventor
Reinhard Becker
Annke Frick
Peter Boderke
Christiane Fürst
Werner Müller
Katrin Tertsch
Ulrich Werner
Petra Loos
Isabell Schöttle
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sanofi SA
Original Assignee
Sanofi SA
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=44148980&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110301081(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US13/110,568 priority Critical patent/US20110301081A1/en
Application filed by Sanofi SA filed Critical Sanofi SA
Assigned to SANOFI reassignment SANOFI ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BODERKE, PETER, FURST, CHRISTIANE, FRICK, ANNKE, MULLER, WERNER, TERTSCH, KATRIN, WERNER, ULRICH, BECKER, REINHARD, LOOS, PETRA, SCHOTTLE, ISABELL
Assigned to SANOFI reassignment SANOFI CORRECTIVE ASSIGNMENT TO CORRECT THE 9TH INVENTOR'S EXECUTION DATE PREVIOUSLY RECORDED ON REEL 026785 FRAME 0991. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: BODERKE, PETER, FURST, CHRISTIANE, FRICK, ANNKE, MULLER, WERNER, TERTSCH, KATRIN, WERNER, ULRICH, BECKER, REINHARD, LOOS, PETRA, SCHOTTLE, ISABELL
Priority to US13/310,118 priority patent/US20120122774A1/en
Publication of US20110301081A1 publication Critical patent/US20110301081A1/en
Priority to US14/220,562 priority patent/US9345750B2/en
Priority to US14/624,575 priority patent/US20150164999A1/en
Priority to US15/134,152 priority patent/US20160228516A1/en
Priority to US15/162,563 priority patent/US20160339084A1/en
Priority to US16/108,064 priority patent/US20190046616A1/en
Priority to US16/368,201 priority patent/US20190388511A1/en
Priority to US16/572,850 priority patent/US20200215163A1/en
Priority to US15/930,643 priority patent/US20200384087A1/en
Priority to US17/176,442 priority patent/US20210308225A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • 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
    • A61K38/2278Vasoactive intestinal peptide [VIP]; Related peptides (e.g. Exendin)
    • 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
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • 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
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin

Definitions

  • the application relates to an aqueous pharmaceutical formulation comprising 200-1000 U/mL [equimolar to 200-1000 IU human insulin] of insulin glargine, and its use.
  • Insulin glargine is 31 B -32 B -Di-Arg human insulin, an analogue of human insulin, with further substitution of asparagine in position A21 by glycine.
  • Lantus® is an insulin product containing insulin glargine providing 24 hour basal insulin supply after single dose subcutaneous injection.
  • Lantus® The glucodynamic effect of Lantus® is distinguished from other currently marketed insulin products by virtue of a delayed and predictable absorption of insulin glargine from the subcutaneous injection site resulting in a smooth, 24 hour time-concentration and action profile without a definite peak.
  • Lantus® was developed to meet the medical need for a long-acting insulin product that can be administered as a single daily injection to yield normal or near-normal blood glucose control with a basal insulin profile that is as smooth as possible over a 24-hour period. Such a preparation provides good control of blood glucose all day, while minimizing the tendency to produce hypoglycemia seen with other insulin preparations with a more definite “peak” effect.
  • a formulation containing 300 U insulin glargine per mL has been developed.
  • the invention is not limited to an insulin glargine U 300 formulation, the clinical studies described herein were performed with an insulin glargine U 300 formulation; each mL insulin glargine U300 contains 300 U (10.9134 mg) insulin glargine. This formulation would allow patients to inject the same number of units of insulin glargine at one third the volume of injection.
  • FIG. 1 Glucose Infusion Rate (GIR) Lantus U100
  • FIG. 2 Glucose Infusion Rate (GIR) Lantus U300
  • FIG. 3 Serum Insulin Concentrations; Lantus U100 (left) and U300 (right)
  • FIG. 4 Blood Glucose (1/2)
  • FIG. 5 Blood Glucose (2/2)
  • FIG. 6 Results of a randomized, 4-sequence, cross-over, double-blind, dose response study of 0.4, 0.6 and 0.9 U/kg HOE-90′-U300 (insulin glargine U300) compared to 0.4 U/kg Lantus® U100 (insulin glargine U100) in patients with diabetes mellitus type 1 using the euglycemic clamp technique.
  • Upper panel insulin glargine concentration (mU/L)
  • middle panel blood glucose (BG, mg/dL)
  • lower panel glucose infusion rate (GIR, mg ⁇ kg ⁇ 1 ⁇ min ⁇ 1 ).
  • the curves display LOWESS smoothed averages of all data points of all subjects (population averages);
  • LOWESS is a data analysis technique for producing a “smooth” set of values from a time series which has been contaminated with noise, or from a scatter plot with a “noisy” relationship between the 2 variables
  • FIG. 7 Glucose infusion rate (GIR, mg ⁇ kg ⁇ 1 ⁇ min ⁇ 1 ).
  • the curves display LOWESS smoothed averages of all data points of all subjects (population averages);
  • LOWESS is a data analysis technique for producing a “smooth” set of values from a time series which has been contaminated with noise, or from a scatter plot with a “noisy” relationship between the 2 variables
  • FIG. 8 Optical microscope pictures of precipitates of insulin glargine formulations with increasing concentrations:
  • A 100 U/mL
  • B 300 U/mL
  • C 500 U/mL
  • D 700 U/mL
  • E 1000 U/mL
  • FIG. 9 Time-action profile of insulin glargine U-100 vs. U-300 in normoglycemic dogs
  • a dose of 0.4 U/kg was selected for this study; it corresponds to the average basal insulin dose in patients. In non-diabetic healthy subjects this dose produces a sizeable elevation in plasma insulin concentration and a lasting glucose lowering effect that can be quantified in euglycemic clamp settings.
  • the replicate design favored by guidelines requires two replicate single dose injections of either IP (R: Lantus® U100, T: insulin glargine U300) in predefined four way cross-over sequences (RTTR or TRRT) as allotted by the randomization plan. This was executed in Periods (P) 1-4 at four different days. As a result, each subject received two replicate single subcutaneous doses of 0.4 U/kg Lantus® U100(R) and insulin glargine U300 (T), alternating between two opposite sites of the periumbilical area.
  • IP Lantus® U100
  • T insulin glargine U300
  • the length of the wash-out period varied individually allowing both the participant and the Investigator to adjust to their needs.
  • 4 days comprise a minimum period for recovery, enabling 1 clamp per week for a participant, while 18 days represent a break of 3 weeks between clamp days, allowing subjects more freedom to fulfill non-study related obligations.
  • the primary objective of the study was to assess the average bioequivalence (ABE) of Lantus® U100 (commercial formulation) and insulin glargine U300 in bioavailability (exposure) and bioefficacy (activity) using the euglycemic clamp technique.
  • ABE bioequivalence
  • the secondary objective of the study was to assess safety and tolerability of insulin glargine U300.
  • both insulin glargine formulations, U100 and U300 were expected to provide the same insulin exposure and the same effectiveness. However, surprisingly insulin exposure and effectiveness were shown to be not the same. Insulin glargine U 100 and insulin glargine U 300 are not equivalent in bio-availability (exposure) and bio-efficacy (activity). Exposure and activity after administration of insulin glargine U300 were less by about 40% as compared to exposure and activity after administration of the same amount (0.4 U/kg) from insulin glargine U100.
  • Insulin glargine U300 did, however, show an even flatter PK (exposure) and PD (activity) profile than insulin glargine U100, as would be desired for a basal insulin. These surprising and unexpected differences in exposure and activity between insulin glargine U100 and insulin glargine U300 formulations after the same s.c. dose to healthy subjects are effectively shown in the figures below. Of note, at the same time blood glucose was constant.
  • the blood glucose lowering effect of insulin glargine was additionally evaluated in healthy, normoglycemic Beagle dogs. With increasing insulin glargine concentration the mean time of action increased from 6.8 h (U100) to 7.69 h (U300), respectively. By increasing the glargine concentration from 100 to 300 U/mL the blood glucose decreasing time-action profile was changed towards a flatter and prolonged activity in the dog.
  • the current data in dogs is consistent with data in humans showing that higher drug concentrations of insulin glargine are positively correlated with profile and longer duration of action.
  • the supernatant of the precipitated insulin was investigated using HPLC technique to determine the insulin glargine content.
  • the present invention is not limited to an insulin glargine U 300 formulation and is effective with other higher concentrated formulations of insulin glargine as outlined in detail in the specification, the clinical studies described herein were performed with an insulin glargine U 300 formulation.
  • the insulin glargine formulations of the present invention exhibit a flatter PK (exposure) and flatter PD (activity) profile than insulin glargine U100 and surprisingly act as improved basal insulins compared to U100 glargine insulin and therefore impart extended duration of exposure and reduce the incidence of hypoglycemia in the treatment of Type I and Type II diabetes, for example.
  • 1 mL of insulin glargine U 300 formulation contains 10.913 mg 21 A -Gly-30 B a-L-Arg-30 B b-L-Arg human insulin [equimolar to 300 IU human insulin], 90 ⁇ g zinc, 2.7 mg m-cresol, 20 mg glycerol 85%, HCl and NaOH ad pH 4.0; specific gravity 1.006 g/mL
  • the pharmaceutical formulation of the present invention contains 200-1000 U/mL of insulin glargine [equimolar to 200-1000 IU human insulin], preferably 250-500 U/mL of insulin glargine [equimolar to 250-500 IU human insulin], more preferred 270-330 U/mL of insulin glargine [equimolar to 270-330 IU human insulin], and even more preferred 300 U/mL of insulin glargine [equimolar to 300 IU human insulin].
  • the present invention is directed to an aqueous pharmaceutical formulation comprising insulin glargine in the range of 200-1000 U/mL [equimolar to 200-1000 IU human insulin], preferably 200 U/ml to 650 U/mL, still preferably 700 U/mL to 1000 U/ml, more preferably 270-330 U/mL and most preferably in a concentration of 300 U/mL.
  • the present invention is directed to an aqueous formulation comprising 200-1000 U/mL [equimolar to 200 to 1000/U human insulin] of insulin glargine, with the proviso that the concentration of insulin in said formulation is not 684 U/ml of insulin glargine.
  • the pharmaceutical formulation of the present invention contains 200 U/mL of insulin glargine (equimolar to 200 IU human insulin] or 300 U/mL of insulin glargine [equimolar to 300 IU human insulin] or 400 U/mL of insulin glargine [equimolar to 400 IU human insulin] or 500 U/mL of insulin glargine [equimolar to 500 IU human insulin] or 600 U/mL of insulin glargine [equimolar to 600 IU human insulin] or 700 U/mL of insulin glargine [equimolar to 700 IU human insulin] or 800 U/mL, of insulin glargine [equimolar to 800 IU human insulin] or 900 U/mL of insulin glargine [equimolar to 900 IU human insulin] or 1000 U/mL of insulin glargine [equimolar to 1000 IU human insulin].
  • Surfactants can be added to pharmaceutical formulation, for example, inter alia, non-ionic surfactants.
  • pharmaceutically customary surfactants are preferred, such as, for example:
  • the surfactants are present in the pharmaceutical composition in a concentration of 5-200 ⁇ g/ml, preferably of 5-120 ⁇ g/ml and particularly preferably of 20-75 ⁇ g/ml.
  • the formulation of the present invention can additionally contain preservatives (e.g. phenol, m-cresol, p-cresol, parabens), isotonic agents (e.g. mannitol, sorbitol, lactose, dextrose, trehalose, sodium chloride, glycerol), buffer substances, salts, acids and alkalis and also further excipients. These substances can in each case be present individually or alternatively as mixtures.
  • preservatives e.g. phenol, m-cresol, p-cresol, parabens
  • isotonic agents e.g. mannitol, sorbitol, lactose, dextrose, trehalose, sodium chloride, glycerol
  • buffer substances e.g. mannitol, sorbitol, lactose, dextrose, trehalose, sodium chloride, glycerol
  • salts e.g., sodium
  • Glycerol, dextrose, lactose, sorbitol and mannitol can be present in the pharmaceutical preparation in a concentration of 100-250 mM, NaCl in a concentration of up to 150 mM.
  • Buffer substances such as, for example, phosphate, acetate, citrate, arginine, glycylglycine or TRIS (i.e. 2-amino-2-hydroxymethyl-1,3-propanediol) buffer and corresponding salts, are present in a concentration of 5-250 mM, preferably 10-100 mM.
  • Further excipients can be, inter alia, salts or arginine.
  • the zinc concentration of the formulation of the present invention is in the range of the concentration which is reached by the presence of 0-1000 ⁇ g/mL, preferably 20-400 ⁇ g/mL zinc, most preferably 90 ⁇ g/mL.
  • the zinc may be present in form of zinc chloride, but the salt is not limited to be zinc chloride.
  • glycerol and/or mannitol can be present in a concentration of 100-250 mmol/L, and/or NaCl is preferably present in a concentration of up to 150 mmol/L.
  • a buffer substance in the pharmaceutical formulation can be present in a concentration of 5-250 mmol/L.
  • a further subject of the invention is a pharmaceutical insulin formulation which contains further additives such as, for example, salts which delay the release of insulin. Mixtures of such delayed-release insulins with formulations described above are included therein.
  • the ingredients are dissolved in water and the pH is adjusted by using HCl and/or NaOH; and likewise by methods known in the art.
  • a further subject of the invention is directed to the use of such formulations for the treatment of diabetes mellitus.
  • a further subject of the invention is directed to the use or the addition of surfactants as stabilizers during the process for the production of insulin, insulin analogs or insulin derivatives or their preparations.
  • the invention further relates to a formulation as described above which additionally comprises also a glucagon-like peptide-1 (GLP1) or an analogue or derivative thereof, or exendin-3 or -4 or an analogue or derivative thereof, preferably exendin-4.
  • GLP1 glucagon-like peptide-1
  • the invention further relates to a formulation as described above in which an analogue of exendin-4 is selected from a group comprising
  • the invention further relates to a formulation as described above in which an analogue of exendin-4 is selected from a group comprising
  • the invention further relates to a formulation as described in the preceding paragraph, in which the peptide-Lys 6 -NH 2 is attached to the C termini of the analogues of exendin-4.
  • the invention further relates to a formulation as described above in which an analogue of exendin-4 is selected from a group comprising
  • the invention further relates to a formulation as described above which additionally comprises Arg 34 , Lys 26 (N ⁇ ( ⁇ -glutamyl(N ⁇ -hexadecanoyl))) GLP-1 (7-37) [liraglutide] or a pharmacologically tolerable salt thereof.
  • the formulation of the present invention can also comprise an analogue of exendin-4, such, for example, lixisentatide, exenatide and liraglutide.
  • exendin-4 analogues are present in the formulation in the range of 0.1 ⁇ g to 10 ⁇ g per U insulin glargine, preferably 0.2 to 1 ⁇ g per U insulin glargine, and more preferably 0.25 ⁇ g to 0.7 ⁇ g per U insulin glargine. Lixisenatide is preferred.
  • the aqueous pharmaceutical formulation can comprise one or more excipients selected from a group comprising zinc, m-cresol, glycerol, polysorbate 20 and sodium.
  • the aqueous pharmaceutical formulation can comprise 90 ⁇ g/mL zinc, 2.7 mg/mL m-cresol and 20 mg/ml glycerol 85%.
  • the aqueous pharmaceutical formulation can comprise 20 ⁇ g/mL polysorbate 20.
  • the pH of the aqueous pharmaceutical formulation is between 3.4 and 4.6, preferably 4 or 4.5.
  • the present invention is directed to a method of treating Type I and Type II Diabetes Mellitus comprising administering to said patient the aqueous pharmaceutical composition of the present invention to a diabetic patient.
  • Preferred among the various disclosed concentration ranges is a concentration of 300 U/mL and the preferred insulin analogue is insulin glargine.
  • the aqueous pharmaceutical formulation also can comprise zinc, m-cresol, glycerol, polysorbate 20 and sodium and mixtures thereof in the ranges disclosed herein in relation to the aqueous pharmaceutical formulation of the present invention.
  • the aqueous pharmaceutical formulation also comprises 0.1 ⁇ g to 10 ⁇ g lixisenatide per U insulin glargine.
  • the insulin is administered preferably once daily but can be administered twice daily as needed. Dosage requirements are a function of the needs of the individual patient determined by the achievement of normal or acceptable blood glucose levels.
  • the present invention is also directed to a method of extending the duration of exposure of insulin glargine in the treatment of Type I and Type II Diabetes Mellitus in a patient comprising administering to said patient the aqueous pharmaceutical formulation of the present invention.
  • Preferred among the various disclosed concentration ranges is a concentration of 300 U/mL.
  • the aqueous pharmaceutical formulation also can comprise zinc, m-cresol, glycerol, polysorbate 20 and sodium and mixtures thereof in the ranges disclosed herein in relation to the aqueous pharmaceutical formulation of the present invention.
  • the aqueous pharmaceutical formulation also comprises 0.1 ⁇ g to 10 ⁇ g lixisenatide per U insulin glargine.
  • the present invention is also directed to a method of reducing the incidence of hypoglycemia in the treatment of Type I and Type II Diabetes Mellitus in a patient with insulin glargine comprising administering to said patient the aqueous pharmaceutical formulation of the present invention.
  • Preferred among the various disclosed concentration ranges is a concentration of 300 U/mL.
  • the aqueous pharmaceutical formulation also can comprise zinc, m-cresol, glycerol, polysorbate 20 and sodium and mixtures thereof in the ranges disclosed herein in relation to the aqueous pharmaceutical formulation of the present invention.
  • the aqueous pharmaceutical formulation also comprises 0.1 ⁇ g to 10 ⁇ g lixisenatide per U insulin glargine.
  • the present invention is also directed to a method of providing a peakless long acting basal insulin in the treatment of Type I and Type II Diabetes Mellitus in a patient with comprising administering to said patient the aqueous pharmaceutical formulation of the present invention.
  • Preferred among the various disclosed concentration ranges is a concentration of 300 U/mL.
  • the aqueous pharmaceutical formulation also can comprise zinc, m-cresol, glycerol, polysorbate 20 and sodium and mixtures thereof in the ranges disclosed herein in relation to the aqueous pharmaceutical formulation of the present invention.
  • the aqueous pharmaceutical formulation also comprises 0.1 ⁇ g to 10 ⁇ g lixisenatide per U insulin glargine.
  • This study was a single center, randomized, controlled, single-blind, four-period, 2-treatment, 2-sequence crossover study in healthy subjects with six visits:
  • the study medication was administered with a replicate of treatment R and T in 2 sequences, RTTR or TRRT at P1 to P4.
  • a washout period of 4 to 18 days was separated each dosing day.
  • P1 must take place no more than 3 to 21 days after SCR. EOS visit must take place between 4 to 14 days after P4.
  • the protocol was submitted to independent ethics committees and/or institutional review boards for review and written approval.
  • the protocol complied with recommendations of the 18th World Health Congress (Helsinki, 1964) and all applicable amendments.
  • the protocol also complied with the laws and regulations, as well as any applicable guidelines, of Germany, where the study was conducted. Informed consent was obtained prior to the conduct of any study-related procedures.
  • Drug code HOE901 (Lantus ® U100 commercial formulation) (Insulin glargine U300 formulation)
  • INN Insulin glargine (recombinant human insulin Insulin glargine (recombinant human insulin analogue) analogue)
  • Formulation Cartridges for 3 mL solution U100 Cartridges for 3 mL solution U300 (1 mL contains 3.637 mg 21 A -Gly-30 B a-L- (1 mL contains 10.913 mg 21 A -Gly-30 B a-L- Arg-30 B b-L-Arg human insulin [equimolar to Arg-30 B b-L-Arg human insulin [equimolar 100 IU human insulin], 30 ⁇ g zinc, 2.7 mg m- to 300 IU human insulin], 90 ⁇ g zinc, 2.7 cresol, 20 mg glycerol 85%, HCl and NaOH mg m-cresol, 20 mg glycerol 85%, HCl and ad pH
  • the body weight recorded during Period 1 Day 1 has been used for calculation of study medication dose for Periods 2, 3 and 4, unless the body weight changed by more than 2 kg compared to Period 1.
  • the amount in Units has been the same for both insulin glargine U100 and insulin glargine U300. This specific gravity is the same for both drug products. However, given the three times higher concentration of insulin glargine in insulin glargine U300 as compared to insulin glargine U100, the to be injected volume and hence the weight has been 1/3 for insulin glargine U300.
  • the syringes providing the individual dose have been prepared by weight. The net weight has been documented only in the source-documentation of the Investigator.
  • Glucose solution 20% glucose solution has been infused with the Biostator to keep subjects individual blood glucose at the determined target level.
  • a second infusion pump (part of the Biostator) has delivered 0.9% sodium chloride solution to keep the line patent. In case the amount of 20% glucose solution needed exceeds the infusion capacity of the Biostator, a second glucose infusion pump has been engaged.
  • Heparin 10000 IU heparin in 100 mL 0.9% sodium chloride solution have been infused into the double lumen catheter at a rate of approximately 2 mL/h to keep it patent for blood glucose measurement by the Biostator.
  • the study medication has been administered only to subjects included in this study following the procedures set out in the clinical study protocol.
  • a randomization schedule has been generated, which has linked the randomization numbers, stratified by gender, to the treatment sequences of the two Lantus® formulations to be injected at P1 to P4.
  • the eligible subjects were randomized by the site.
  • the randomization number was allocated to the subject number subsequently in the order in which subjects' eligibility has been confirmed before P1.
  • the first subject for a gender stratum qualifying after SCR received the first randomization number for the appropriate gender stratum.
  • the next subject who qualifies within a stratum received the next randomization number within the stratum.
  • the randomization number has been used as the treatment kit number to allocate the treatment kit to the subject.
  • Each subject were given the study medication carrying the treatment kit number to which he has been allocated to.
  • the treatment kit containing the IP carried general information, treatment kit number, period number, a field to write the subject number on the container-box, and additional statements as required by local regulations.
  • Subjects who permanently discontinue from the study retained subject number and randomization number, if already given.
  • the study medication has been packed by Sanofi-Aventis GmbH, Frankfurt am Main, Germany according to the randomization plan.
  • the cartridges containing the study medication and the cartons they were packed in have been labeled with the study number, the randomization number, batch number, storage conditions, Sponsor and the P number.
  • the Pharmacist or the person designated by him Before study medication administration, the Pharmacist or the person designated by him has prepared the syringes with the appropriate study medication and has labeled the syringe with the subject number, the randomization number and the appropriate period according to the study medication containers.
  • the content of the labeling was in accordance with the local regulatory specifications and requirements.
  • the study medication was stored protected from light at a temperature of +2° C. to +8° C.
  • the study medication was prevented from freezing. During preparation it was not necessary to have the medication protected from light.
  • Stimulation of insulin receptors by insulin glargine is the mode of action.
  • Subsequent peripheral glucose uptake and suppression of endogenous glucose production comprise the glucodynamic effects producing a reduction in blood glucose concentration.
  • the resulting glucose utilization is best characterized by the gauge of glucose required to keep the blood glucose concentration constant.
  • the euglycemic clamp technique has been employed to assess the amount of glucose needed to keep blood glucose concentrations at 5% below baseline level after injection of insulin glargine.
  • Offsite blood glucose has been determined with a Super GL glucose analyzer also using the glucose oxidase method.
  • the amount of glucose utilized per unit (dose) of subcutaneously injected insulin is a measure of the glucodynamic effect.
  • the continuously recorded glucose infusion rate is a reflection of the time action profile of the injected insulin.
  • the primary pharmacodynamic variable is the area under the glucose infusion rate time curve within 24 hours [GIR-AUC 0-24 h (mg ⁇ kg ⁇ 1 )].
  • the secondary pharmacodynamic variable is the time to 50% GIR-AUC 0-24 h [T 50% -GIR-AUC (0-24 h) (h)].
  • Blood samples for assessment of serum insulin glargine and C-peptide concentrations have been taken 1 hour, 30 min and immediately prior to subcutaneous injection of study medication, thereafter 30 min, 1 hour, 2 hours and then bi-hourly up to 24 hours, and 30 hours after injection.
  • the numbering of insulin glargine samples was P00, P01, P02, P03, P04, etc.
  • the numbering of C-peptide samples was C00, C01, C02, C03, C04, etc (see also study flow chart).
  • Serum insulin glargine concentrations have been determined using a radioimmunoassay (RIA) for human insulin (Insulin RIA kit, ADALTIS, Italy) calibrated for insulin glargine. Kit REF 10624.
  • RIA radioimmunoassay
  • LLOQ lower limit of quantification
  • Serum C-peptide concentrations have been determined using a radioimmunoassay (RIA) for C-peptide (C-peptide RIA kit, ADALTIS, Italy). Kit REF C-peptide 10282.
  • RIA radioimmunoassay
  • the lower limit of quantification (LLOQ) was 0.090 nmol/L.
  • the insulin glargine concentration time curve was a measure of the systemic insulin exposure of subcutaneously injected IP.
  • the primary pharmacokinetic variable was the area under the serum insulin glargine concentration time curve [INS-AUC 0-24 h ( ⁇ U ⁇ h ⁇ mL ⁇ 1 )].
  • the secondary pharmacokinetic variable was the time to 50% INS-AUC 0-24 h [T 50% -INS-AUC (0-24 h) (h)].
  • Each study period (P1 to P4) lasted 2 days, Day 1 and Day 2.
  • Day 1 was the starting day of the euglycemic clamp and administration of study medication.
  • Day 2 was day of the end of the euglycemic clamp, which lasted 30 hours after study medication administration.
  • No strenuous activity e.g. mountain biking, heavy gardening etc.
  • Consumption of alcoholic beverages, grapefruit juice, and stimulating beverages containing xanthine derivatives (tea, chocolate, coffee, CokeTM-like drinks, etc.) and grapefruit has not been permitted from 24 hours before until completion of the euglycemic clamp.
  • the subjects have fasted (except for water) for 10 hours before Day 1 of each study period (P1 to P4) and remained fasting (except for water) until end of the euglycemic clamp.
  • the subjects had to stay in the clinic for approximately 32 hours at each clamp visit.
  • the 9-digit subject number has been allocated to the subject, starting with 276001001.
  • the next subject who qualifies to enter SCR has received the subject number 276001002 etc.
  • the first subject has received the randomization number 101.
  • the next subject who qualifies has received the randomization number 102.
  • the blood pressure, heart rate and core body temperature have been recorded in supine position after at least 5 minutes rest in the morning of Day 1, prior to and after completion of clamp procedures 30 hours after each study medication administration (Day 2).
  • Body weight, alcohol screen and RBC, Hb, HcT (only before clamp period of P3 and P4) have been assessed only before starting the clamp in the morning of Day 1.
  • a dorsal hand vein or lateral wrist vein of the left arm has been cannulized in retrograde fashion and connected to a Biostator (Life Sciences instruments, Elkhart, Ind., USA) in order to continuously draw arterialized venous blood for the determination of blood glucose.
  • a Biostator Life Sciences instruments, Elkhart, Ind., USA
  • a second venous line has been placed into the antecubital vein of the left arm and have been used to collect samples for serum insulin glargine and reference blood glucose determination.
  • a third vein has been cannulised on the contralateral forearm allowing the infusion of 20% glucose solution and 0.9% saline with the Biostator.
  • the Biostator determined blood glucose levels and adjusted the glucose infusion rate to maintain blood glucose levels at 5% below the individual fasting blood glucose, determined as the mean of the 3 fasting blood glucose values measured 60, 30 and 5 minutes before study medication administration. Additional blood samples of 0.3 mL for the determination of blood glucose have been taken 60, 30, and 5 minutes before administration of the study medication to check against a laboratory reference based on the glucose oxidase method.
  • insulin glargine U100 commercial formulation
  • insulin glargine U300 have been injected in the periumbilical area 5 cm lateral to the umbilicus (left, right, left, right) using a standardized skin fold technique.
  • U100 insulin syringes manufactured by Beckton & Dickinson
  • 0.5 mL volume with a needle of 0.30 mm ⁇ 8 mm (30 G) have been used.
  • the study medication was labeled with their respective treatment kit number subject number (to be documented on the container-box after randomization), and Period number see Section 8.5 Packaging and Labeling).
  • Blood samples for determination of serum insulin glargine and C-peptide concentrations have been taken 1 hour, 30 min and immediately before medication and thereafter 30 min, 1 hour, 2 hours and then bi-hourly up to 24 hours, and 30 hours after administration of study medication.
  • Injection sites have been observed during the entire clamp period. Any changes in the health condition of the subjects have been reported in the subject's medical records (source) and the CRF.
  • RBC, Hb and Hct at P 3 have been analyzed for incurring anemia at P 4. If positive, the interval between P 3 and P 4 have been extended to the maximum allowed 18 days and an additional RBC, Hb and Hct assessment made prior to P 4.
  • Subjects have returned for an EOS visit between 4 to 14 days after P4. Subjects have fasted (apart from water) for 10 hours. Any changes in the health condition of the subjects since the last period have been reported in the subject's medical records (source) and the CRF.
  • Urine drug screen consists of amphetamines/metamphetamines, barbiturates, benzodiazepines, cannabinoids, cocaine, opiates.
  • Qualitative safety urinalysis with dipsticks have been conducted at SCR and EOS.
  • Safety urinalysis consists of analysis for: pH, protein, glucose, blood, erythrocytes, leukocytes, bilirubin, urobilinogen, ketone, specific gravity, and nitrite
  • Blood pressure and heart rate have been measured after about 5 minutes rest in a supine position, and also after 3 minutes in an upright position at SCR and EOS.
  • blood pressure and heart rate have been recorded in supine position after at least 5 minutes prior to start of clamp procedures in the morning of day 1, and after completion of clamp procedures 30 hours after each study medication administration (day 2).
  • Electrocardiograms (standard 12-lead) have been recorded at SCR and EOS.
  • Body weight and height have been measured at SCR. The body weight have been recorded in the morning of Day 1 of P1 to P4 (prior to administration of study medication) and at EOS. Alcohol screen (ethanol, breath analyzer) have been conducted at SCR and EOS, and in the morning of Day 1 of P1 to P4 (prior to administration of study medication).
  • the CRF have been considered as source documentation for other items.
  • This example provides information for the statistical analysis plan for the study.
  • a statistical analysis plan have been drafted prior to inclusion of subjects.
  • INS-AUC (0-24 h) have been the primary parameter for which therefore the sample size calculation was performed.
  • sample size calculation For the purpose of this sample size calculation, several within-subject SD within of natural log-transformed INS-AUC (0-24 h) between 0.125 and 0.225 were considered.
  • a sample size calculation method for an average bioequivalence approach was used for a 4-period, 2-treatment, 2-sequence cross-over design. If the 90% CIs for the formulation ratio have been wholly contained within [0.80-1.25], then average bioequivalence have been concluded for the parameter.
  • Study HOE901/1022 was the basis for assumptions on variability. Based on the statistical analysis of study HOE901/1022, a value of 0.175 could be expected for the within subject standard deviation (SD within ) on the natural log-transformed scale.
  • a number of 24 randomized subjects accounts for potential cases of withdrawals.
  • Subject disposition at the final visit have been presented in a listing including sequence group, disposition status at the end of the study with the date of last administration of study drug, date of final visit, reason for discontinuation. All withdrawals from the study, taking place on or after the start of the first study drug administration, have been fully documented in the body of the clinical study report (CSR).
  • CSR clinical study report
  • Protocol deviations have been identified by the study team before database lock and listed in the Data Review Report, including missing data and IP discontinuations, and classified as minor or major deviations.
  • Safety evaluation have been based on subjects who received a dose of study drug (exposed population), regardless of the amount of treatment administered, including subjects prematurely withdrawn.
  • BMI Body mass index
  • BMI body weight[kg] ⁇ (height[m]) ⁇ 2
  • the latest scheduled value before study drug administration within the period or within the study, whatever is applicable for the variable, have been taken as the baseline value. If the baseline pre-dosing value is rechecked before dosing, the rechecked value have been considered as the baseline and used in statistics.
  • GIR glucose infusion rate
  • GIR max In order to provide meaningful and reliable data, the value for GIR max and correspondingly the time to GIR max have been derived from a smoothed GIR curve for each subject.
  • the mixed model includes fixed terms for sequence, period, formulation, and random terms for subject within sequence, with formulation specific between-subject and within-subject variances and subject-by-formulation variance. Point estimate and 90% confidence interval for the formulation ratio (T/R) have then been obtained based on Fieller's theorem [Fieller, 1954].
  • Formulation ratios (T/R) with confidence limits have been derived for fractional GIR-AUCs (mg ⁇ kg ⁇ 1 ) and maximum standardized glucose infusion rate [GIR max (mg ⁇ kg ⁇ 1 ⁇ min ⁇ 1 )] using the corresponding linear mixed effects model as described for the primary analysis.
  • the individual on-treatment phase for analysis of safety data have started with the first administration of study medication and has ended with the EOS visit.
  • each TEAE has been assigned to the last formulation given before onset and/or worsening of the AE. If a TEAE develops on one formulation and worsens under a later formulation, it has been considered a TEAE for both formulations.
  • an AE has been counted as a TEAE, unless it can clearly be ruled out that it is not a TEAE (e.g. by partial dates or other information).
  • start date of an AE is incomplete or missing, it has been assumed to have occurred after the first administration of study medication except if an incomplete date indicated that the AE started prior to treatment.
  • PCSA Potentially clinically significant abnormalities
  • out-of-range criteria have been defined in the statistical analysis plan of this study. Definitions of potentially clinically significant abnormalities (PCSA) and out-of-range definitions have been reported by parameter.
  • Subjects with values out of normal ranges and subjects with PCSAs have been analyzed by formulation, and overall for end of study evaluation. Subjects with post-baseline PCSAs have been listed.
  • PCSA Potentially clinically significant abnormalities
  • out-of-range criteria have been defined in the statistical analysis plan of this study. Definitions of PCSA and out-of-range definitions have been reported by parameter.
  • Subjects with PCSAs have been analyzed by formulation, and overall for end of study evaluation. Subjects with post-baseline PCSAs have been listed.
  • PCSA Potentially clinically significant abnormalities
  • out-of-range criteria have been defined in the statistical analysis plan of this study. Definitions of PCSA and out-of-range definitions have been reported by parameter.
  • Serum insulin concentrations have been individually listed and descriptive statistics per time point have been generated.
  • the mixed model included fixed terms for sequence, period, formulation, and random terms for subject within sequence, with formulation specific between-subject and within-subject variances and subject-by-formulation variance.
  • PK/PD analyses have been performed in an explorative manner, if appropriate.
  • PD pharmacodynamic
  • PK pharmacokinetic
  • BMI Body mass indexes
  • the area under the serum insulin glargine concentration time curve from 0 to 24 hours was not equivalent for Lantus U 100 and Lantus U 300.
  • the exposure was less by about 40% with U300.
  • the area under the GIR versus time curve from 0 to 24 hours was not equivalent for Lantus U 100 and Lantus U 300.
  • the activity was less by about 40% with U300.
  • the time to 50% of INS-AUC (0-24 h) (h) was similar for Lantus U 100 and Lantus U 300.
  • the time to 50% of GIR-AUC (0-24 h) (h) was greater by 0.545 (h) (0.158-1.030) for Lantus U 300, which was statistically significant.
  • Insulin glargine U 100 and insulin glargine U 300 are not equivalent in bio-availability (exposure) and bio-efficacy (activity). Exposure and activity after insulin glargine U300 were less by about 40% as compared to exposure and activity after administration of the same amount (0.4 U/kg) from insulin glargine U100.
  • Insulin glargine U300 did, however, show an even flatter PK (exposure) and PD (activity) profile than insulin glargine. U100, as would be desired for a basal insulin. These surprising and unexpected differences in exposure and activity between insulin glargine U100 and insulin glargine U300 formulations after the same s.c. dose to healthy subjects are effectively shown in the figures below. Of note, at the same time blood glucose was constant.
  • results from the study in healthy subjects showed the inequivalence in exposure and effectiveness between Lantus® U100 and insulin glargine U300.
  • Subjects received the same dose of insulin glargine (0.4 U/kg) for U100 and U300, but delivery of the same unit-amount from U300 produced about 40% less exposure and effect than delivery from U100.
  • Insulin glargine U300 did, however, show an even flatter pharmacodynamic profile than Lantus® U100, as would be desired for a basal insulin.
  • a new study described in the following examples therefore compares the glucodynamic activity and exposure of three different subcutaneous doses of insulin glargine U300 versus a standard dose of Lantus® U100 as comparator in a euglycemic clamp setting with type 1 diabetes patients.
  • This study aims to approximate an U300 dose that is equieffective to 0.4 U/kg Lantus® U100 as assessed by parameters of blood glucose disposal provided by the clamp technique.
  • Insulin glargine exposure is assessed from concentration-time profiles after subcutaneous administration and activity as glucose utilization per unit insulin.
  • the study is designed to assess the metabolic effect and exposure of different insulin glargine U300 doses compared to a standard dose of Lantus® U100 in a euglycemic clamp setting in subjects with diabetes mellitus type 1.
  • the study comprises 4 treatments (R, T 1 , T 2 and T 3 ), 4 treatment periods (TP1-4) and 4 sequences.
  • Subjects are exposed to each treatment R, T 1 , T 2 and T 3 once in a cross-over, double-blind and randomized manner according to a Latin square design. This design is considered appropriate to evaluate the pharmacological effect and exposure of different insulin glargine U300 doses compared to Lantus® U100.
  • the Lantus® U100 dose of 0.4 U/kg selected for the study is well characterized to provide euglycemia in type 1 diabetes patients and has been readily investigated in other clamp studies with type 1 diabetes patients.
  • insulin glargine U300 Three different doses are tested for insulin glargine U300, 0.4, 0.6 and 0.9 U/kg. This dose range allows intrapolating an approximate dose equieffective to 0.4 U/kg Lantus® U100.
  • the dose of 0.4 U/kg of insulin glargine U300 has already been tested in healthy volunteers (see examples 1-6) and was found to be less active than 0.4 U/kg Lantus® U100 within 30 hours, the predefined end of observation period. Bioactivity of 0.4 U/kg insulin glargine U300 as measured by the total glucose disposition was 39.4% lower than that of reference medication (0.4 U/kg Lantus® U100).
  • a correspondingly higher dose of insulin glargine U300 e.g.
  • the active pharmaceutical ingredient, insulin glargine is the same in both formulations, U100 and U300.
  • the doses used in this study are within the range of regular use. Although an overall risk of hypoglycemia is not completely excluded, it is controlled by the euglycemic clamp technique.
  • the pharmacodynamic activity of insulin glargine is evaluated by the euglycemic clamp technique in type 1 diabetes patients, which is the established standard procedure to evaluate the effect of exogenous administered insulin products on blood glucose disposal.
  • GIR body weight standardized glucose infusion rate
  • total glucose disposed GIR-AUC 0-36
  • times to a given percentage of GIR-AUC 0-36 such as time to 50% of GIR-AUC 0-36 .
  • Ancillary parameters are the maximum smoothed body weight standardized GIR, GIR max , and Time to GIR max , GIR-T max .
  • Duration of action of insulin glargine is derived from the time between dosing and pre-specified deviations above the euglycemic (clamp) level.
  • Glucose monitoring is performed for 36 hours due to the long duration of action of insulin glargine after subcutaneous administration
  • T 50% INS-AUC 0-36 the time to 50% of INS-AUC (T 50% INS-AUC 0-36 ) is calculated as a measure for the time location of the insulin glargine exposure profile, and INS-C max and INS-T max will serve as additional measures.
  • the primary objective of the study is to assess the metabolic effect ratios of three different insulin glargine U300 doses versus 0.4 U/kg Lantus® U100.
  • the secondary objectives of the study are to assess the exposure ratios of three different insulin glargine U300 doses versus 0.4 U/kg Lantus® U100, to compare the duration of action of different insulin glargine U300 doses versus 0.4 U/kg Lantus® U100, to explore the dose response and dose exposure relationship of insulin glargine U300, and to asses the safety and tolerability of insulin glargine U300 in subjects with type 1 diabetes.
  • Phase I single-center, double-blind, randomized, cross-over (4 treatments, 4 treatment periods and 4 sequences; Latin square), active control, with a wash-out duration between treatment periods (5-18 days, preferred 7 days) in male and female subjects with type 1 diabetes mellitus receiving single-doses of insulin glargine at
  • the four treatments R and T 1-3 are given cross-over in four treatment periods (TP 1 to TP 4) with the four-sequences
  • Number of subjects planned At least 24 subjects are to be enrolled to have 20 evaluable subjects.
  • Injections are given left or right of the umbilicus, with both sites being used for separate injections.
  • a washout period of 5 to 18 days separates consecutive dosing days, the preference is 7 days (7 days between consecutive dosing).
  • the length of the wash-out period varies individually allowing both the participant and the investigator to adjust to their needs.
  • 5 days comprise a minimum period for recovery enabling 1 clamp per week for a participant, while 18 days represent a break of 3 weeks between dosing days, allowing subjects the freedom to fulfill non-study related obligations, if unavoidable.
  • IP administration is administered under fasting conditions; subject continues to fasten throughout the whole clamp period.
  • the blood glucose concentration is within a range of 5.5 mmol/L (100 mg/dL) ⁇ 20% without any glucose infusion for the last hour prior to dosing during pre-clamp.
  • IP is administered. IP administration does not occur earlier than 09:00 clock time in the morning and not later than 14:00 clock time on Day 1 in Treatment Periods 1 to 4. If blood glucose is not stabilized before 14:00 hours, dosing does not occur. The visit is terminated and the subject is scheduled for a new dosing visit 1-7 days later.
  • IP administration is done by a person who is not otherwise involved in the study or part of the study team at the CRO. This person gets the random code to prepare IP administration in accordance to the open random list and doses subjects accordingly. The preparation and dosing is followed and checked by a second independent person. Respective documents of dose preparation and treatment sequence is kept strictly confidential and is not being disclosed to any other person.
  • the body weight (in kg) is determined to one decimal place and the amount of insulin calculated is rounded up or down to integer numbers as shown in the following examples for a dose of 0.6 U/kg insulin glargine:
  • the body weight recorded during TP1 D1 is used for calculation of study medication dose for all treatment periods.
  • the study medication dose is not to be changed if a subject's weight changes by less than or equal to 2 kg between TP1 and one of the subsequent TPs. If a subject's body weight changes by more than 2 kg between TP1 and one of the subsequent TPs, the study medication dose is re-calculated based on the weight at D1 of the respective treatment period.
  • Syringes with needles attached appropriate to accurately administer small amounts of injection solution are used only (e.g. Becton Dickinson, Ref 305502, Dimensions: 1 mL 27 G 3 ⁇ 80.40 ⁇ 10).
  • the syringes are supplied by the investigator.
  • Glucose solution sodium chloride solution, heparin and insulin glulisine is provided by the Investigator.
  • Glucose solution 20% glucose solution is infused with the BiostatorTM to keep subjects individual blood glucose at the determined target level.
  • a second infusion pump (part of the BiostatorTM) delivers 0.9% sodium chloride solution to keep the line patent. In case the amount of 20% glucose solution needed exceeds the infusion capacity of the BiostatorTM, a second glucose infusion pump is engaged.
  • Heparin A low dose heparin solution (10.000 Units heparine/100 mL saline) is infused via a double lumen catheter. The heparin solution is taken up together with blood used for the Biostator'sTM blood glucose measurement in the other lumen of the catheter and is aimed to prevent blood clotting in the system.
  • Insulin glulisine 15 U Apidra® [100 U/mL] is given to 49 mL of saline solution, to which 1 mL of the subject's own blood is added to prevent adhesion, producing a concentration of 0.3 U/mL, which is infused at an individual rate to achieve euglycemia.
  • Subjects receive four different treatments (R, T 1 , T 2 and T 3 ) in a randomized, blinded and crossover design.
  • IP dispensing and administration In order to maintain the blinding, a third party un-blinded person is involved for IP dispensing and administration. This person is not otherwise involved in the study and/or part of the study team at the CRO, does not disclose any information to anyone and ensures to maintain blinding condition of the study, He/she gets the random code and doses subjects accordingly. The preparation of IP and dosing is followed and checked by a second independent person who has also access to the random code but is equally bound to confidentiality.
  • IPs are administered according to the Clinical Study Protocol only to subjects who have given written informed consent.
  • IP administration is in accordance with the randomized treatment sequence.
  • Subjects withdrawn from the study retain their subject number and their treatment number, if already assigned. Replacement subjects have a different identification number (i.e., 500+the number of the subject who discontinued the study). Each subject receives the same treatment sequence as the subject, who discontinued the trial
  • Screen Failed subjects are assigned a different number, e.g., 901, 902 (to be recorded in the CRF only in case of AE occurring during screening period after signing of informed consent).
  • Baseline parameters are the parameters available the closest before the dosing.
  • Insulin glargine U300 solution is provided by sanofi-aventis in regrouping boxes of 3 mL cartridges.
  • IP The respective number of IP is packaged under the responsibility of sanofi-aventis according to good manufacturing practice and local regulatory requirement and provided to CRO.
  • the content of the labeling is in accordance with the local regulatory specifications and requirements.
  • Lantus® U100 is commercially available and will be ordered by the CRO.
  • the IP has to be stored at +2° C. to +8° C., protected from light, and must not be frozen.
  • a third party un-blinded person is responsible for IP dispensing and administration. This person is not otherwise involved in the study and/or part of the study team at the CRO, does not disclose any information to anyone and ensures to maintain blinding condition of the study. He/she gets the random code and doses subjects accordingly. The preparation of IP and dosing is followed and checked by a second independent person who has also access to the random code but is equally bound to confidentiality.
  • code-breaking material which contains the name of the treatment is supplied as envelopes. It is kept in a safe place on site throughout the Clinical Trial. The Sponsor retrieves all code-breaking material (opened or sealed) on completion of the Clinical Trial.
  • the Investigator documents the date of opening and reason for code breaking in the source data.
  • the Investigator, the clinical site pharmacist, or other personnel allowed to store and dispense IP is responsible for ensuring that the IP used in the study is securely maintained as specified by the Sponsor and in accordance with the applicable regulatory requirements.
  • IP is dispensed in accordance with the Clinical Trial Protocol and it is the Investigator's responsibility to ensure that an accurate record of IP issued and returned is maintained.
  • the last subcutaneous injection of short-acting insulin is no later than 9 hours before study drug administration. Subjects on pump therapy discontinues the insulin infusion in the morning of Day 1, at least 6 hours prior to each IP administration (around 03:00 clock time assuming start of IP administration at 09:00).
  • acetaminophen/paracematol is prohibited if there is a known risk of hepatotoxicity, or as soon as abnormalities of liver enzymes occur.
  • the present study is designed to assess the metabolic effect and exposure ratios of three different insulin glargine U300 doses versus 0.4 U/kg Lantus® U100, to compare the duration of action of different insulin glargine U300 doses versus 0.4 U/kg Lantus® U100, to explore the dose response and dose exposure relationship of insulin glargine U300, and to asses the safety and tolerability of insulin glargine U300 in an euglycemic clamp setting in subjects with diabetes mellitus type 1.
  • the pharmacodynamic effect of insulin glargine mainly the total glucose disposal and duration of insulin action, is evaluated by the euglycemic clamp technique.
  • the amount of glucose required is a measure of the glucose uptake into tissues (glucose disposal or glucose lowering activity) mediated by the exogenous insulin excess.
  • the BiostatorTM determines blood glucose levels in 1 min intervals and adjusts the glucose infusion rate in response to changes in blood glucose using a predefined algorithm.
  • the last subcutaneous injection of short-acting insulin is no later than 9 hours before IP administration.
  • Subjects on pump therapy discontinue the insulin infusion in the morning of Day 1, at least 6 hours prior to each IP administration (around 03:00 clock time assuming start of IP administration at 09:00).
  • Treatment Periods 1 to 4 (TP1-TP4), subjects are admitted to the clinic in the morning of D1 after an overnight fast of at least 10 hours.
  • blood glucose is then adjusted to 5.5 mmol/L (100 mg/dL) ⁇ 20% (euglycemic clamp level) without any glucose infusion for the last hour prior to dosing.
  • the insulin glulisine infusion is discontinued immediately prior to the administration of the study medication.
  • Subjects receive reference or test medication (R, T 1-3 , see Table 4) as assigned by randomization. Injections is given left or right of the umbilicus.
  • IP administration does not occur earlier than 09:00 clock time in the morning and not later than 14:00 clock time on Day 1 in Treatment Periods 1 to 4. If blood glucose is not stabilized during pre-clamp before 14:00 clock time, dosing does not occur. The visit is terminated and the subject is scheduled for a new dosing visit 1-7 days later.
  • IP administration is administered under fasting conditions; subject continues to fasten throughout the whole clamp period.
  • the euglycemic clamp blood glucose level is continuously maintained by means of iv infusion of glucose solution until clamp end.
  • any basal insulin supplementation is to add to or even to substitute endogenous insulin secretion between meals.
  • exogenous insulin should provide for just the amount of insulin required to dispose hepatic glucose production. If perfectly matched, there is no need for extra glucose to compensate for excess insulin. The resulting glucose infusion rate approximates zero. Once insulin action ceases, blood glucose concentration rises. The times to onset of rise and to times blood glucose concentrations exceeding predefined thresholds are read by the BiostatorTM.
  • Selected doses of Lantus® U100 and insulin glargine U300 are above the average basal need which in turn produce some glucose demand reflected in a sizeable GIR up to 36 hours.
  • the corresponding parameter indicative of the clamp performance i.e. the precision for keeping blood glucose at clamp baseline level, is the blood glucose variability over the clamp period.
  • a measure for blood glucose variability is the coefficient of variation (CV %) per individual clamp.
  • a low coefficient of variation in blood glucose is a prerequisite to properly assess the insulin effect in clamp settings.
  • the clamp period is not to exceed 36 hours post study medication injection, the predefined clamp end.
  • insulin glulisine used in the pre-IP administration time of the clamp is given to extend the observation period to 36 hours. In that case, the sponsor has to be informed.
  • the subjects are delinked from the clamp setting when blood glucose is well within the isoglycemic range.
  • IPs The effect of the IPs is to last about 24-36 hours, which is why the participants is confined to the institute for 2 days.
  • a washout period of 5 to 18 days separates consecutive clamp period days, the preference is 7 days (7 days between consecutive dosing).
  • the length of the wash-out period varies individually allowing both the participant and the investigator to adjust to their needs.
  • 5 days comprise a minimum period for recovery enabling 1 clamp per week for a participant, while 18 days represent a break of 3 weeks between dosing days, allowing subjects the freedom to fulfill non-study related obligations, if unavoidable.
  • Arterialized venous blood is continuously drawn at a rate of 2 mL/h for determination of arterial blood glucose concentration every minute during pre-clamp (prior to IP administration) and clamp period (up to 36 hours after IP administration).
  • Blood glucose is continuously measured during the clamp procedure.
  • at least 74 samples per subject and treatment period will be collected for calibration of the BiostatorTM after IP administration.
  • 74*4*24 samples or 7104 samples are collected (see table below).
  • Duration of blood glucose control is taken as the time in euglycemia from dosing to deviation above clamp glucose level (100 mg/dL). Times of controlled blood glucose within predefined margins is taken from dosing to specified thresholds, e.g. blood glucose levels at 110, 130 and 150 mg/dL.
  • GIR max the maximum smoothed body weight corrected GIR
  • GIR-T max the time to GIR max
  • ECGs Twelve-lead ECGs are recorded after at least 10 minutes in supine position using an electrocardiographic device (MAC 550TM).
  • the electrodes are positioned at the same place for each ECG recording throughout the study (attachment sites of the leads are marked with an indelible pen).
  • Findings at the site of injection are graded mainly according to a Global Irritation Score.
  • a local injection site reaction with a score of ⁇ 3 according to the rating scale is documented additionally as an adverse event.
  • the subjects are asked to report sensations at the injection site.
  • the area under the insulin concentration curve (INS-AUC) up to 36 hours, INS-AUC 0-36 and the time to 50% of INS-AUC 0-36 is derived.
  • the maximum insulin concentration INS-C max , and time to C max (INS-T max ) is obtained.
  • Blood is collected for the determination of insulin glargine concentrations at time points 0H, 1H, 2H, 4H, 6H, 8H, 12H, 16H, 20H, 24H, 28H, 32H and 36H after injection of study medication.
  • the following pharmacokinetic parameters are calculated, using non-compartmental methods for insulin glargine concentrations after single dose.
  • the parameters include, but are not be limited to the following.
  • a third party un-blinded person is involved for IP dispensing and administration. This person is not otherwise involved in the study and/or part of the study team at the CRO or sponsor. He/she gets the random code provided by sanofi-aventis and does not disclose the random code or any other information to any other person. For safety reason, the treatment randomization code is unblinded for reporting to the Health Authority of any Suspected Unexpected Adverse Drug Reaction (SUSAR) and reasonably associated with the use of the IP according to either the judgment of the Investigator and/or the Sponsor.
  • SUSAR Suspected Unexpected Adverse Drug Reaction
  • the Investigator is the primary person responsible for taking all clinically relevant decisions in case of safety issues.
  • a specialist should be envisaged in a timely manner (e.g. acute kidney failure, convulsions, skin rashes, angioedema, cardiac arrest, electrocardiographic modifications, etc).
  • Screening procedures are carried out within 28 days up to 3 days prior to inclusion to determine subject's eligibility for participation.
  • the subject receives information on the study objectives and procedures from the investigator.
  • the subject signs the informed consent prior to any action related to the study. Recording of adverse events starts thereafter.
  • the screening visit includes the following investigations:
  • Subjects, who qualify for enrollment into the study, are admitted to the clinic in the fasted state in the morning of D1 of TP1 at approximately 07:00.
  • the inclusion examination is carried out on the first dosing day (D1, TP1) and includes the following investigations:
  • Each subject receives an incremental identification number according to the chronological order of his/her inclusion in the study.
  • Randomization occurs on D1/TP1 after confirmation of subject's eligibility by the Investigator. If more than one subject is randomized at the same time, subjects are randomized consecutively according to the chronological order of inclusion on the morning of Day 1/TP1, i.e. the subject with the lowest subject number receives the next available randomization number.
  • Results of laboratory tests of D1/TP1 are baseline values and considered confirmatory, with the exception of the ⁇ -HCG urine test (based on sample collected during screening visit), which must be negative.
  • a blood sample is taken for archiving and for determination of anti-insulin antibodies (on D1/TP1 only).
  • the last subcutaneous injection of short-acting insulin is no later than 9 hours before IP administration.
  • Subjects on pump therapy discontinues the insulin infusion in the morning of Day 1, at least 6 hours prior to each IP administration (around 03:00 clock time assuming start of IP administration at 09:00).
  • the amount of insulin glargine required for injection will be calculated according to subject's body weight.
  • Hematology is analyzed for incurring anemia on Day 1 of Treatment Period 3. If positive, the wash-out interval between Treatment Periods 3 and 4 is extended to the maximum allowed 18 days or start of TP4 will be postponed until hematological parameters have been normalized. An additional hematological assessment is made on Day 1 of Treatment Period 4.
  • Subjects are then be prepared for the start of the pre-clamp procedure with three venous lines connected to an automatic glucose reading device (BiostatorTM) and remain in semi-recumbent position for the entire duration of the sampling period.
  • BiostatorTM automatic glucose reading device
  • a dorsal hand vein or lateral wrist vein of the left arm is cannulated and connected to the BiostatorTM in order to continuously draw arterialized venous blood for the determination of blood glucose concentration.
  • the left hand is placed into a heated box (“Hot-Box”), which provides for an air temperature of about 55° C., allowing arterialization of venous blood.
  • a second venous line is placed into the antecubital vein of the left arm and is used to collect samples for insulin and reference blood glucose determination.
  • a third vein is cannulated on the contralateral forearm allowing the infusion of 0.9% saline and 20% glucose solution with a pump in the BiostatorTM or insulin glulisine with an external pump.
  • the blood glucose level is maintained within 4.4 to 6.6 mmol/L (80-120 mg/dL, pre-clamp).
  • additional intravenous bolus injection of insulin glulisine is given to keep the blood glucose within the target range.
  • no intravenous bolus injections are given until clamp end.
  • Additional blood samples for the determination of blood glucose are taken in at least 30 min intervals to check against a laboratory reference based on the glucose oxidase method. If necessary the BiostatorTM is re-calibrated according to results of the laboratory reference method.
  • Insulin infusion rates are adjusted individually. While keeping blood glucose at the target level both, insulin and glucose infusion rate are minimized during the clamp run-in phase.
  • Insulin glulisine solution is infused by means of a high precision infusion pump (Terumo Spritzenpumpe TE 311TM), 20% glucose solution is be applied by a high precision infusion pump (Terumo Infusionspumpe TE 171TM).
  • the clamp level is adjusted 60 min before study medication administration to maintain the blood glucose at about 5.5 mmol/L (100 mg/dL) until the end of the clamp period.
  • the pre-clamp is prolonged and IP administration postponed until 14:00 clock time in case the target glucose level has not been met during the run-in phase (pre-clamp). If the target glucose level cannot be established within until 14:00 clock time, the visit is terminated and the subject may be scheduled for a new dosing visit 1-7 days later.
  • the insulin glulisine infusion is discontinued immediately before study medication administration.
  • the first insulin sample for PK is taken immediately thereafter.
  • the study medication is administered (Table 4), either
  • the study medication is administered preferably by the same person at during the whole study.
  • the end of the injection defines time zero (T0), which defines the starting time of the subsequent clamp period and PK sampling.
  • Every clamp observation period lasts 36 hours and thus ends at approximately at 21:00 on D2, the predefined end-of-clamp. Thereafter the subjects are delinked from the euglycemic clamp setting when blood glucose is well within the isoglycemic range, receive a meal and their usual insulin treatment.
  • the rapid acting insulin analog e.g. insulin glulisine
  • the rapid acting insulin analog used in the pre-IP administration time of the clamp is given to extend the clamp period to 36 hours for pharmacokinetic blood sampling.
  • the sponsor has to be informed. Thereafter the subjects are delinked from the euglycemic clamp setting when blood glucose is well within the isoglycemic range, receive a meal and their usual insulin treatment.
  • the injection site reaction is assessed 15 minutes as well as one hour after injection of the study medication and documented as an AE if a score of ⁇ 3 is observed according to the rating scale.
  • EOS end-of-study
  • the Investigator ensures that based on all available clinical results, the subject can be safely released from the study.
  • Subjects ceases their usual insulin treatment on Days ⁇ 2 to ⁇ 1, depending on the type of insulin used (long acting, NPH, intermediate). Thereafter, the blood glucose levels are controlled solely by multiple subcutaneous injections of the usual short-acting insulin.
  • the subjects do not take any concomitant medication, which will interfere with the metabolic control or the insulin sensitivity of subjects throughout the study and in the two weeks before the study.
  • Consumption of alcoholic beverages, grapefruit juice, and stimulating beverages containing xanthine derivatives is not permitted 24 hours before administration of each study medication until the end of the clamp.
  • Orange juice or similar carbohydrates are given as corrective measures for hypoglycemia during clamp if not adequately counteracted by intravenous glucose infusion when connected to the BiostatorTM.
  • Subjects who smoke 5 or less cigarettes per day are included in the study and subjects may smoke during the study, except on D1 and D2 of TP1 to TP4.
  • Fluid supply is at least 2500 mL for each 36-hour period.
  • the primary objective of the study is to assess the relative metabolic effect for insulin glargine given as one dose of U100 (R) and three different doses of U300 (T 1 to T 3 ). Based on the data of study PKD10086, a value of approximately 0.375 can be expected for the SD within of GIR-AUC end of clamp on the natural log-transformed scale.
  • Table 11 shows the maximum imprecision (in terms of the 90% confidence interval) for a pairwise treatment ratio of adjusted geometric means that will be obtained with 90% assurance, for total number of subject N between 16 and 24, assuming a true within-subject SD of values between 0.325 and 0.425 for log GIR-AUC 0-36 .
  • Subject disposition at the final visit is presented in a listing including sequence group, disposition status at the end of the study with the date of last administration of study drug, date of final visit, reason for discontinuation. All withdrawals from the study, taking place on or after the start of the first study drug administration, are fully documented in the body of the clinical study report (CSR).
  • CSR clinical study report
  • analyses are conducted according to the treatment received rather than according to the randomized treatment.
  • the bioanalytical assay for insulin glargine is interfered by other insulins like insulin glulisine. Therefore, the pharmacokinetic data for insulin glargine of those subjects are excluded from evaluation, who have received (for safety reasons) insulin glulisine within the clamp observation period of 36 hours after IP administration.
  • BMI Baseline body mass index
  • the latest scheduled value before study drug administration within the period or within the study, whatever is applicable for the variable, is taken as the baseline value. If the baseline pre-dosing value is rechecked before dosing, the rechecked value is considered as the baseline and used in statistics.
  • Prior and concomitant medications/therapies are coded according to the World Health Organization-Drug Reference List (WHO-DRL, latest version in use at time of database lock) and are listed individually.
  • WHO-DRL World Health Organization-Drug Reference List
  • Insulin infusion or bolus given at any time during the clamp procedure is listed or plotted over time on an individual basis.
  • Insulin infusion or bolus given after dosing during the clamp procedure is listed on an individual basis.
  • the blood glucose concentration and glucose infusion rate (GIR) is continuously recorded during the clamp procedure.
  • GIR glucose infusion rate
  • the following PD variable is considered primary.
  • GIR-AUC 0-36 is calculated according to the rectangular rule for the stepwise constant function with timescale in minutes.
  • the maximum of the raw body weight standardized GIR is subject to the noise in the GIR adjustment.
  • the derivation of GIR max and the time to GIR max is based upon a LOESS (locally weighted regression in smoothing scatterplots) smoothing technique for the raw body weight standardized GIR data. Due to the expected morphology of the GIR-profiles as known under Lantus®, a smoothing factor of 6% is used (SAS®, PROC LOESS, factor 0.06).
  • LOESS locally weighted regression in smoothing scatterplots
  • GIR-AUC 0-36 is log-transformed (natural log).
  • PD parameters are listed individually, and descriptive statistics are generated by treatment.
  • Treatment ratios (T 1 /R, T 2 /R, T 3 /R) with confidence limits are derived for maximum standardized glucose infusion rate [GIR max (mg*min/kg)] using the corresponding linear mixed effects model as described above for the primary analysis. Exploratory comparisons between treatments are based on conventional bioequivalence criteria (90% confidence limits 0.80 to 1.25).
  • GIR-T max values The distribution of GIR-T max values is represented by histogram plots for each treatment. In addition, a histogram of differences in GIR-T max between test treatments and reference is provided.
  • T 50% -GIR-AUC 0-36 (h) is analyzed non-parametrically based on Hodges-Lehmann method for paired treatment comparisons, CIs for pair-wise treatment differences (T1-R, T2-R, T3-R) in medians are derived.
  • the distribution of T 50% -GIR-AUC 0-36 values is represented by histogram plots for each treatment.
  • a histogram of differences in T 50% -GIR-AUC 0-36 between treatments (T1-R, T2-R, T3-R) is provided.
  • GIR-T max values The distribution of GIR-T max values is represented by histogram plots for each treatment. In addition, a histogram of differences in GIR-T max between test treatments and reference is provided.
  • Duration of clamp is derived per clamp as the time between dosing and end of clamp in hours.
  • Individual variability of blood glucose per clamp is derived as the coefficient of variation (CV %) of blood glucose values between individual start and individual end of clamp (or first administration of insulin glulisine during clamp).
  • Individual average blood glucose level per clamp is derived as the arithmetic mean of blood glucose values between individual start and individual end of clamp (or first administration of insulin glulisine during clamp).
  • the safety evaluation is based upon the review of the individual values (potentially clinically significant abnormalities), descriptive statistics (summary tables, graphics) and if needed on statistical analysis (appropriate estimations, confidence intervals).
  • “Potentially Clinically Significant Abnormalities” (PCSA) criteria are used according to standard criteria of sanofi-aventis. Criteria are documented in the statistical analysis plan of this study. The safety analysis is conducted according to the sanofi-aventis standards related to analysis and reporting of safety data from clinical trials.
  • the observation period is divided into segments of three different types:
  • each TEAE is assigned to the last treatment given before onset (or worsening) of the AE. If a TEAE develops on one treatment and worsens under a later treatment, it is considered treatment emergent for both treatments.
  • an AE is counted as a TEAE, unless it can clearly be ruled out that it is not a TEAE (e.g. by partial dates or other information).
  • start date of an AE is incomplete or missing, it is assumed to have occurred after the first administration of study medication except if an incomplete date indicates that the AE started prior to treatment.
  • the values to be used as baseline are the values collected on D1 predose in the first treatment period. If any of the scheduled baseline tests are repeated for any subject, the last rechecked values are considered as baselines, provided they were done before the first IP administration.
  • liver function data CPK
  • eosinophils are expressed as multiple of the corresponding ULN.
  • Heart rate and systolic and diastolic blood pressure are measured after 10 minutes in supine resting position and also after 3 minutes in standing position, except when connected to the BiostatorTM.
  • the values to be used as the baselines are the D1 pre-dose assessment value of each treatment period. If any of the scheduled baseline tests are repeated for any subject, the last rechecked values are considered as baselines, provided they were done before the IP administration.
  • orthostatic differences are calculated as the change from supine to standing position.
  • the values to be used as baselines for body weight and BMI are the values collected on D1 of TP1.
  • the values to be used as baselines for body temperature are the values collected on D1 of each TP.
  • Heart rate, PQ-, QRS-, and QT-intervals and corrected QT (QTc) from automatic reading are analyzed as raw parameter value and change from baseline.
  • the values to be used as the baseline are the Day 1 predose value of each period. If any of the scheduled baseline tests are repeated for any subject, the rechecked values are considered as baselines, provided they were done before the drug administration of the period.
  • an on-treatment analysis is performed using all post-baseline assessments done during the on-treatment period, including rechecked values. Counts of subjects with postbaseline PCSAs are provided in summary tables regardless of the normal or abnormal status of the baseline, by treatment group.
  • Raw data for all parameters and change from baseline are summarized in descriptive statistics by parameter, treatment, and time of measurement.
  • a listing of individual data from subjects with post-baseline PCSAs is provided, sorted by type of measurement and sorted by subject, period, and time of measurement.
  • a separate listing of the cardiac profile for subjects with prolonged QTc >450 ms for Males and >470 ms for Females) or changes from baseline in QTc>60 ms (for males and females) and a listing of subjects with at least one abnormality in qualitative assessment (i.e., abnormal ECG) after the 1st dosing are also provided.
  • Frequency distributions by treatment are provided for levels of local tolerability at injection site. Individual data are listed. Within each criterion and treatment, a subject is counted with their most severe result.
  • Allergic medical history and family medical history is documented for subjects with any occurrence of potential allergic reaction. All details of allergic medical history and of allergic family medical history are listed on an individual basis.
  • a summary table is provided with the number of subjects for the anti-insulin antibodies results during the study and from the post study investigations. Individual subject listing is provided.
  • T 50% -AUC 0-36 for insulin is derived in the context of the statistical analysis.
  • Pharmacokinetic parameters of insulin glargine are listed and summarized using at least arithmetic and geometric means, standard deviation (SD), standard error of the mean (SEM), coefficient of variation (CV %), minimum, median and maximum for each treatment.
  • SD standard deviation
  • SEM standard error of the mean
  • CV coefficient of variation
  • AUC 0-36 for insulin glargine. Prior to all analysis described below, AUC 0-36 values are log-transformed (natural log).
  • Estimate and 90% confidence interval (CI) for the ratio of treatments geometric means (T 1 /R, T 2 /R, T 3 /R) are obtained by computing estimate and 90% CI for the difference between treatment means within the linear mixed effects model framework, and then converting to ratio of geometric means by the antilog transformation. Bioequivalence is concluded if the 90% CI for the ratio is entirely within the 0.80 to 1.25 equivalence reference interval.
  • T 50% -AUC 0-36 values for insulin is represented by histogram plots for each treatment.
  • a histogram of differences in T 50% -AUC 0-36 between treatments (T 1 -R, T 2 -R, T 3 -R) is provided.
  • T 50% -AUC 0-36 (h) is analyzed non-parametrically.
  • the empirical power model provides a readily and interpretable measure of the degree of non-proportionality, which can be used both to confirm proportionality and to assess the pharmacokinetic and clinical significance of any departures.
  • the analysis of dose proportionality studies requires estimation rather than significance testing in order that the pharmacokinetic and clinical significance of any non-proportionality can be assessed.
  • the power model is fit on the log-transformed scale using a random coefficients power model for dose (in U/kg body weight):
  • log(parameter) (log(alpha)+alpha[ i ])+(beta+beta[ i ])*log(dose)
  • log(alpha) and beta are the population intercept and slope, respectively, and alpha[i] and beta[i] are the random deviations from alpha and beta, respectively, for the i-th subject.
  • Estimates for beta with 90% confidence intervals are obtained via estimated generalized least squares in the SAS®/PROC MIXED procedure, with restricted maximum likelihood (REML) estimates of covariance parameters.
  • the mixed effect model (as used for analysis of treatment ratios) is used for the analysis.
  • Estimates with 90% CIs for the parameter increases associated with pairwise dose increases are obtained by first computing estimates with CIs for pairwise differences between doses in the mixed effects model framework, and then converting to ratios using the antilog transformation.
  • a total of 24 subjects with Type 1 diabetes mellitus were enrolled, randomized and received at least one dose of study medication. Of the 24 randomized subjects, 2 subject withdrew from the study on own request. Twenty-two (22) subjects completed the study according to the protocol and were included in the pharmacodynamic (PD) and pharmacokinetic (PK) analyses. All 24 treated subjects were included in the safety evaluation.
  • PD pharmacodynamic
  • PK pharmacokinetic
  • BMI Body mass indexes
  • BMI body weight[kg] ⁇ (height[m]) ⁇ 2 .
  • the area under the serum insulin glargine concentration time curve from 0 to 36 hours was not equivalent for R and T1 and T2 and about equivalent with T3.
  • the exposure was estimated to be less by about 37% with T1, less by about 43% with T2 and similar with T3, compared to R.
  • the area under the GIR versus time curve from 0 to 36 hours was not equivalent for R and T1 and T2 and about equivalent with T3.
  • the exogenous glucose consumption required to preserve blood glucose control was estimated to be less by about 88% with T1, 67% with T2 while about similar with T3.
  • the time to 50% of INS-AUC (0-36 h) (h) with R was about 14 h and thus shorter as compared to about 16 h, 16 h and 19 h with T1, T2 and T3, respectively.
  • the time to 50% of GIR-AUC (0-36 h) (h) with R was about 12 h and thus shorter as compared to about 17 h, 18 h and 20 h with T1, T2 and 13, respectively.
  • T1, T2 and T3 did, however, show yet flatter PK (exposure) and PD (activity) profiles with even less fluctuation around the averages than R, i.e., a profile as it would be desired for basal insulin supply. This is particularly evident when comparing R and T3 which provide nominal equivalent total exposure and total glucose consumption though of different profiles.
  • results from the study in healthy subjects and in subjects with Type 1 diabetes mellitus showed exposure and effectiveness not to be equivalent between Lantus® U100 and insulin glargine U300.
  • HOE901-U300 did, however, show even less fluctuation in exposure and pharmacodynamic profiles, as it would be desired for basal insulin supply, with a yet even longer duration of action.
  • a new study described in the following examples therefore compares two different subcutaneous doses of insulin glargine U300 versus a standard dose of Lanais® U100 as comparator with a final euglycemic clamp setting in patients with type 1 diabetes mellitus.
  • This study aims to estimate an U300 dose that is equieffective to 0.4 U/kg Lantus® U100 as assessed by parameters of blood glucose control and blood glucose disposal provided by the clamp technique.
  • Insulin glargine exposure is assessed from concentration-time profiles after repeated subcutaneous administration at steady state, and activity as glucose utilization per unit insulin at steady state.
  • the study comprises two cross-over treatments (R and T1, and R and T2) in 2 parallel groups, with 2 treatment periods (TP1, TP2) and 2 sequences, each.
  • the Lantus® U100 dose of 0.4 U/kg selected for the study is well characterized to provide euglycaemic blood glucose control in type 1 diabetes patients and has been readily investigated in other clamp studies with type 1 diabetes patients.
  • insulin glargine U300 Two different doses are tested for insulin glargine U300, 0.4 and 0.6 U/kg. This dose range allows intrapolating an approximate dose equieffective to 0.4 U/kg Lantus® U100.
  • the dose of 0.4 U/kg of insulin glargine U300 has already been tested in healthy volunteers and subjects with type diabetes mellitus (see foregoing examples) and was found to be less active than 0.4 U/kg Lantus® U100 within 30 and 36 hours, respectively, the predefined ends of the observation periods. Blood glucose control with 0.4 U/kg insulin glargine U300 required less total glucose disposition than that of reference medication (0.4 U/kg Lantus® U100).
  • a correspondingly higher dose of insulin glargine U300, e.g. 0.6 U/kg insulin glargine U300 is expected to result in even tighter blood glucose control at less total glucose disposition.
  • the proportional dose escalation allows exploring exposure and effect profiles for dose-proportionality.
  • a study in patients with type 1 diabetes avoids confounding impact of endogenous insulin and better permits assessment of exposure and duration of action.
  • the active pharmaceutical ingredient, insulin glargine is the same in both formulations, U100 and U300.
  • the doses used in this study are within the range of regular use. Although an overall risk of hypoglycemia is not completely excluded, it is controlled by the euglycemic clamp technique.
  • the pharmacodynamic activity of insulin glargine is evaluated by the euglycemic clamp technique in type 1 diabetes patients, which is the established standard procedure to evaluate the effect of exogenous administered insulin products on blood glucose disposal.
  • GIR body weight standardized glucose infusion rate
  • total glucose disposed within 24 and 36 hours respectively
  • GIR-AUC 0-24 and GIR-AUC 0-36 and times to a given percentage of GIR-AUC 0-24 and GIR-AUC 0-36 such as time to 50% of GIR-AUC 0-36 .
  • Ancillary parameters are the maximum smoothed body weight standardized GIR, GIR max , and Time to GIR max , GIR-T max .
  • Duration of action of insulin glargine is derived from the time between dosing and pre-specified deviations above the euglycemic (clamp) level.
  • Glucose monitoring is performed for 36 hours due to the long duration of action of insulin glargine after subcutaneous administration
  • the time to 50% of INS-AUC (e.g. T 50% INS-AUC 0-36 ) is calculated as a measure for the time location of the insulin glargine exposure profile, and INS-C max and INS-T max will serve as additional measures.
  • the primary objective of the study is to assess the blood glucose control and the required exogenous glucose consumption of two different insulin glargine U300 doses versus 0.4 U/kg Lantus® U100 in steady state.
  • the secondary objectives of the study are to assess in steady state, the exposure ratios of two different insulin glargine U300 doses versus 0.4 U/kg Lantus® U100, to compare the duration of action of two different insulin glargine U300 doses versus 0.4 U/kg Lantus® U 100, to explore the dose response and dose exposure relationship of insulin glargine U300, and to asses the safety and tolerability of insulin glargine U300 in subjects with type 1 diabetes.
  • the supernatant of the precipitated insulin is investigated using HPLC technique to determine the insulin glargine content.
  • sodium dihydrogen phosphate monohydrate (M: 137.98 g/mol) are dissolved per mL water.
  • 0.1M sodium hydroxide or 0.1M hydrochloric acid is used for adjustment of the pH to 7.4.
  • Solutions of insulin glargine drug product having concentrations of up to 1000 U/mL and comprising the same total amount of insulin glargine and the buffer are placed in plastic tubes and are slightly shaken. After precipitation of the insulin glargine the dispersions are centrifuged at slow rotations for a pre-defined time period. A defined volume of the dissolution medium is taken out and replaced with fresh buffer medium.
  • the content of insulin glargine in the samples from the supernatant is quantified against the respective insulin reference standard by reverse-phase-HPLC using a two mobile phase system, containing a sodium dihydrogenphosphate buffer in water, sodium chloride (NaCl) and different amounts of acetonitrile.
  • detection wavelength is 215 nm.
  • the release profile of insulin glargine from the higher concentrated solutions is flatter and prolonged compared to Lantus U100.
  • the precipitates of insulin glargine formulations having concentrations of 100 U/mL, 300 U/mL, 500 U/mL 700 U/mL and 1000 U/mL have been investigated by microscopy.
  • Said formulations (with an identical amount of 60 U of insulin glargine) have been precipitated in 200 ⁇ L of a phosphate buffer, pH 7.4 and were investigated by transmitted light optical microscope (Olympus Model BX61) with the magnitudes 100 ⁇ , the pictures are shown in the following also presenting the maximum diameters.
  • the blood glucose lowering effect of insulin glargine was evaluated in healthy, normoglycemic Beagle dogs.
  • the dogs received single subcutaneous injections of 0.3 IU/kg. Venous blood glucose was determined before the first injection and subsequently up to 24 h.
  • Animals were taken from cohort of ⁇ 30 healthy, normoglycemic male Beagle dogs, originally obtained from Harlan. The dogs were maintained in kennel groups under standardized conditions. The day before study start the dogs were randomly distributed to study cages. They were fasted 18 hours prior to start and throughout the experiment with free access to tap water. Body weight of the dogs in the present study was between 13 and 27 kg. After each experiment the dogs were allowed to recover for at least two weeks.
  • Blood sampling was performed consecutively via puncture of the forearm vein (Vena cephalica) before drug administration (0 h) and thereafter up to 24 hours. Blood glucose was determined enzymatically (Gluco-quant® Glucose/HK kit on Roche/Hitachi 912).
  • HbA1c Glycocylated hemoglobin HBs Hepatitis B surface
  • QTc QT interval automatically corrected by the ECG machine QTcB QT interval corrected by Bazett formula QTcF QT interval corrected by Fridericia formula QtcN QT interval corrected by a population approach QtcNi QT interval corrected by individual population approach RBC Red Blood Cell count
US13/110,568 2010-05-19 2011-05-18 Long-acting formulations of insulins Abandoned US20110301081A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
US13/110,568 US20110301081A1 (en) 2010-05-19 2011-05-18 Long-acting formulations of insulins
US13/310,118 US20120122774A1 (en) 2010-05-19 2011-12-02 Long-acting formulations of insulins
US14/220,562 US9345750B2 (en) 2010-05-19 2014-03-20 Long-acting formulations of insulin
US14/624,575 US20150164999A1 (en) 2010-05-19 2015-02-17 Long-Acting Formulations of Insulin
US15/134,152 US20160228516A1 (en) 2010-05-19 2016-04-20 Long-Acting Formulations of Insulin
US15/162,563 US20160339084A1 (en) 2010-05-19 2016-05-23 Long-Acting Formulations of Insulin
US16/108,064 US20190046616A1 (en) 2010-05-19 2018-08-21 Long-Acting Formulations of Insulin
US16/368,201 US20190388511A1 (en) 2010-05-19 2019-03-28 Long-Acting Formulations of Insulin
US16/572,850 US20200215163A1 (en) 2010-05-19 2019-09-17 Long-Acting Formulations of Insulin
US15/930,643 US20200384087A1 (en) 2010-05-19 2020-05-13 Long-Acting Formulations of Insulin
US17/176,442 US20210308225A1 (en) 2010-05-19 2021-02-16 Long-Acting Formulations of Insulin

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
EP10305532 2010-05-19
EP10305532.3 2010-05-19
EP10305780 2010-07-13
EP10305780.8 2010-07-13
US41160810P 2010-11-09 2010-11-09
US201161429936P 2011-01-05 2011-01-05
EP11305140 2011-02-10
EP11305140.3 2011-02-10
US13/110,568 US20110301081A1 (en) 2010-05-19 2011-05-18 Long-acting formulations of insulins

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/310,118 Continuation US20120122774A1 (en) 2010-05-19 2011-12-02 Long-acting formulations of insulins
US14/220,562 Continuation US9345750B2 (en) 2010-05-19 2014-03-20 Long-acting formulations of insulin

Publications (1)

Publication Number Publication Date
US20110301081A1 true US20110301081A1 (en) 2011-12-08

Family

ID=44148980

Family Applications (11)

Application Number Title Priority Date Filing Date
US13/110,568 Abandoned US20110301081A1 (en) 2010-05-19 2011-05-18 Long-acting formulations of insulins
US13/310,118 Abandoned US20120122774A1 (en) 2010-05-19 2011-12-02 Long-acting formulations of insulins
US14/220,562 Active US9345750B2 (en) 2010-05-19 2014-03-20 Long-acting formulations of insulin
US14/624,575 Abandoned US20150164999A1 (en) 2010-05-19 2015-02-17 Long-Acting Formulations of Insulin
US15/134,152 Abandoned US20160228516A1 (en) 2010-05-19 2016-04-20 Long-Acting Formulations of Insulin
US15/162,563 Abandoned US20160339084A1 (en) 2010-05-19 2016-05-23 Long-Acting Formulations of Insulin
US16/108,064 Abandoned US20190046616A1 (en) 2010-05-19 2018-08-21 Long-Acting Formulations of Insulin
US16/368,201 Abandoned US20190388511A1 (en) 2010-05-19 2019-03-28 Long-Acting Formulations of Insulin
US16/572,850 Abandoned US20200215163A1 (en) 2010-05-19 2019-09-17 Long-Acting Formulations of Insulin
US15/930,643 Abandoned US20200384087A1 (en) 2010-05-19 2020-05-13 Long-Acting Formulations of Insulin
US17/176,442 Pending US20210308225A1 (en) 2010-05-19 2021-02-16 Long-Acting Formulations of Insulin

Family Applications After (10)

Application Number Title Priority Date Filing Date
US13/310,118 Abandoned US20120122774A1 (en) 2010-05-19 2011-12-02 Long-acting formulations of insulins
US14/220,562 Active US9345750B2 (en) 2010-05-19 2014-03-20 Long-acting formulations of insulin
US14/624,575 Abandoned US20150164999A1 (en) 2010-05-19 2015-02-17 Long-Acting Formulations of Insulin
US15/134,152 Abandoned US20160228516A1 (en) 2010-05-19 2016-04-20 Long-Acting Formulations of Insulin
US15/162,563 Abandoned US20160339084A1 (en) 2010-05-19 2016-05-23 Long-Acting Formulations of Insulin
US16/108,064 Abandoned US20190046616A1 (en) 2010-05-19 2018-08-21 Long-Acting Formulations of Insulin
US16/368,201 Abandoned US20190388511A1 (en) 2010-05-19 2019-03-28 Long-Acting Formulations of Insulin
US16/572,850 Abandoned US20200215163A1 (en) 2010-05-19 2019-09-17 Long-Acting Formulations of Insulin
US15/930,643 Abandoned US20200384087A1 (en) 2010-05-19 2020-05-13 Long-Acting Formulations of Insulin
US17/176,442 Pending US20210308225A1 (en) 2010-05-19 2021-02-16 Long-Acting Formulations of Insulin

Country Status (43)

Country Link
US (11) US20110301081A1 (ru)
EP (9) EP3636250A1 (ru)
JP (5) JP5269945B2 (ru)
KR (4) KR101486743B1 (ru)
CN (5) CN107583037A (ru)
AR (1) AR081200A1 (ru)
AU (1) AU2011202239C1 (ru)
BR (2) BR122019006884B1 (ru)
CA (1) CA2792669C (ru)
CL (1) CL2012003197A1 (ru)
CO (1) CO6640217A2 (ru)
CR (1) CR20120578A (ru)
CY (3) CY1115649T1 (ru)
DK (3) DK2387989T3 (ru)
DO (1) DOP2012000261A (ru)
EC (1) ECSP12012298A (ru)
ES (4) ES2945208T3 (ru)
GT (1) GT201200287A (ru)
HK (5) HK1248593A1 (ru)
HR (3) HRP20140860T1 (ru)
HU (2) HUE039355T2 (ru)
IL (2) IL221964A (ru)
JO (2) JO2944B1 (ru)
LT (2) LT2781212T (ru)
MA (1) MA34224B1 (ru)
MX (2) MX343489B (ru)
MY (1) MY156188A (ru)
NI (1) NI201200152A (ru)
NZ (1) NZ602541A (ru)
PE (2) PE20160781A1 (ru)
PH (2) PH12015501421B1 (ru)
PL (4) PL2387989T3 (ru)
PT (3) PT2781212T (ru)
RS (1) RS53559B1 (ru)
RU (3) RU2564104C2 (ru)
SG (2) SG10201510406YA (ru)
SI (3) SI2781212T1 (ru)
TN (1) TN2012000478A1 (ru)
TR (2) TR201809475T4 (ru)
TW (4) TWI605825B (ru)
UY (1) UY33391A (ru)
WO (1) WO2011144673A2 (ru)
ZA (1) ZA201206998B (ru)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013158277A1 (en) * 2012-04-18 2013-10-24 Waters Technologies Corporation Methods for quantifying polypeptides using mass spectrometry
WO2016001862A1 (en) 2014-07-04 2016-01-07 Wockhardt Limited Extended release formulations of insulins
US20160030339A1 (en) * 2013-04-03 2016-02-04 Sanofi Treatment of Diabetes Mellitus by Long-Acting Formulations of Insulins
US20160058840A1 (en) * 2013-04-30 2016-03-03 Novo Nordisk A/S Novel Administration Regime
US9345750B2 (en) 2010-05-19 2016-05-24 Sanofi Long-acting formulations of insulin
US9364519B2 (en) 2011-09-01 2016-06-14 Sanofi-Aventis Deutschland Gmbh Pharmaceutical composition for use in the treatment of a neurodegenerative disease
US9408893B2 (en) 2011-08-29 2016-08-09 Sanofi-Aventis Deutschland Gmbh Pharmaceutical combination for use in glycemic control in diabetes type 2 patients
US9526764B2 (en) 2008-10-17 2016-12-27 Sanofi-Aventis Deutschland Gmbh Combination of an insulin and a GLP-1-agonist
US9603904B2 (en) 2008-10-30 2017-03-28 Novo Nordisk A/S Treating diabetes melitus using insulin injections with less than daily injection frequency
US9670261B2 (en) 2012-12-21 2017-06-06 Sanofi Functionalized exendin-4 derivatives
US9694053B2 (en) 2013-12-13 2017-07-04 Sanofi Dual GLP-1/glucagon receptor agonists
US9707176B2 (en) 2009-11-13 2017-07-18 Sanofi-Aventis Deutschland Gmbh Pharmaceutical composition comprising a GLP-1 agonist and methionine
US9751926B2 (en) 2013-12-13 2017-09-05 Sanofi Dual GLP-1/GIP receptor agonists
US9750788B2 (en) 2013-12-13 2017-09-05 Sanofi Non-acylated exendin-4 peptide analogues
US9758561B2 (en) 2014-04-07 2017-09-12 Sanofi Dual GLP-1/glucagon receptor agonists derived from exendin-4
US9771406B2 (en) 2014-04-07 2017-09-26 Sanofi Peptidic dual GLP-1/glucagon receptor agonists derived from exendin-4
US9775904B2 (en) 2014-04-07 2017-10-03 Sanofi Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
US9789165B2 (en) 2013-12-13 2017-10-17 Sanofi Exendin-4 peptide analogues as dual GLP-1/GIP receptor agonists
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
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
US9950039B2 (en) 2014-12-12 2018-04-24 Sanofi-Aventis Deutschland Gmbh Insulin glargine/lixisenatide fixed ratio formulation
US9981013B2 (en) 2010-08-30 2018-05-29 Sanofi-Aventis Deutschland Gmbh Use of AVE0010 for the treatment of diabetes mellitus type 2
US9982029B2 (en) 2015-07-10 2018-05-29 Sanofi Exendin-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
US10029011B2 (en) 2009-11-13 2018-07-24 Sanofi-Aventis Deutschland Gmbh Pharmaceutical composition comprising a GLP-1 agonist, an insulin and methionine
US10159713B2 (en) 2015-03-18 2018-12-25 Sanofi-Aventis Deutschland Gmbh Treatment of type 2 diabetes mellitus patients
US10335464B1 (en) 2018-06-26 2019-07-02 Novo Nordisk A/S Device for titrating basal insulin
US10434147B2 (en) 2015-03-13 2019-10-08 Sanofi-Aventis Deutschland Gmbh Treatment type 2 diabetes mellitus patients
US10596229B2 (en) 2010-10-27 2020-03-24 Novo Nordisk A/S Method of treating diabetes mellitus by administration, at specifically defined intervals, of a derivative of a naturally occurring insulin or insulin analogue, the derivative having a prolonged profile of action
US10758592B2 (en) 2012-10-09 2020-09-01 Sanofi Exendin-4 derivatives as dual GLP1/glucagon agonists
US10806797B2 (en) 2015-06-05 2020-10-20 Sanofi Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate
US11167035B2 (en) 2005-12-28 2021-11-09 Novo Nordisk A/S Insulin compositions and method of making a composition
US20220202711A1 (en) * 2013-04-03 2022-06-30 Sanofi Treatment of Diabetes Mellitus by Long-Acting Formulations of Insulins

Families Citing this family (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2524423C2 (ru) 2008-01-09 2014-07-27 Санофи-Авентис Дойчланд Гмбх Новые производные инсулина с чрезвычайно замедленным профилем время/действие
CN107080836A (zh) * 2009-07-06 2017-08-22 赛诺菲-安万特德国有限公司 含有甲硫氨酸的水性胰岛素制备物
WO2012101253A1 (en) 2011-01-28 2012-08-02 Sanofi Pharmaceutical compositions comprising human antibodies to pcsk9
AR087305A1 (es) 2011-07-28 2014-03-12 Regeneron Pharma Formulaciones estabilizadas que contienen anticuerpos anti-pcsk9, metodo de preparacion y kit
FR2984749A1 (fr) * 2011-12-23 2013-06-28 Adocia Solution injectable a ph 7 comprenant au moins une insuline basale dont le pi est compris entre 5,8 et 8,5 en combinaison avec une hormone gastro-intestinale a visee therapeutique
CA2843586A1 (fr) 2011-08-10 2013-02-14 Adocia Solution injectable d'au moins une insuline basale
FR2985429B1 (fr) * 2012-01-09 2016-07-29 Adocia Solution injectable a ph 7 comprenant au moins une insuline basale dont le pi est compris entre 5,8 et 8,5 et un polyaminoacide substitue obtenu par un procede de polymerisation controle
CN104114155B (zh) * 2012-01-09 2019-02-15 阿道恰公司 Ph为7并且至少包含pi为5.8至8.5之基础胰岛素和经取代共聚(氨基酸)的可注射溶液
FR2985428B1 (fr) * 2012-01-09 2016-05-27 Adocia Solution injectable a ph 7 comprenant au moins une insuline basale dont le pi est compris entre 5,8 et 8,5 et un polyaminoacide substitue
AR092862A1 (es) * 2012-07-25 2015-05-06 Hanmi Pharm Ind Co Ltd Formulacion liquida de insulina de accion prolongada y un peptido insulinotropico y metodo de preparacion
FR3001896B1 (fr) 2013-02-12 2015-07-03 Adocia Solution injectable a ph 7 comprenant au moins une insuline basale dont le point isolectrique est compris entre 5,8 et 8,5 et un polymere anionique hydrophobise
US20150314003A2 (en) 2012-08-09 2015-11-05 Adocia Injectable solution at ph 7 comprising at least one basal insulin the isoelectric point of which is between 5.8 and 8.5 and a hydrophobized anionic polymer
FR3001895B1 (fr) 2013-02-12 2015-07-03 Adocia Solution injectable a ph7 comprenant au moins une insuline basale dont le point isoelectrique est compris en 5,8 et 8,5 et un compose anionique porteur de charges carboxylates et de radicaux hydrophobes
US9707275B2 (en) * 2012-12-19 2017-07-18 Wockhardt Limited Stable aqueous composition comprising human insulin or an analogue or derivative thereof
CN103893744B (zh) * 2012-12-24 2017-12-19 杭州九源基因工程有限公司 一种治疗糖尿病的药物制剂及其制备方法
JP2016505601A (ja) * 2012-12-26 2016-02-25 ウォックハート リミテッド 医薬組成物
TWI641381B (zh) 2013-02-04 2018-11-21 法商賽諾菲公司 胰島素類似物及/或胰島素衍生物之穩定化醫藥調配物
CN105188736A (zh) * 2013-03-20 2015-12-23 诺和诺德股份有限公司 胰岛素给药方案
JP6438944B2 (ja) * 2013-06-17 2018-12-19 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 インスリングラルギン/リキシセナチド固定比率製剤
CN104587455A (zh) * 2013-10-31 2015-05-06 江苏万邦生化医药股份有限公司 一种胰岛素制剂
TW201609800A (zh) 2013-12-13 2016-03-16 賽諾菲公司 做為雙重glp-1/升糖素受體促效劑之艾塞那肽-4胜肽類似物
MX2016008978A (es) 2014-01-09 2016-10-04 Sanofi Sa Formulaciones farmaceuticas de analogos de insulina y/o derivados de insulina estabilizadas y que estan libres de glicerol.
SG11201604706TA (en) 2014-01-09 2016-07-28 Sanofi Sa Stabilized pharmaceutical formulations of insulin aspart
RU2016132342A (ru) 2014-01-09 2018-02-14 Санофи Стабилизированные фармацевтические составы на основе инсулиновых аналогов и/или инсулиновых производных
CN103830189A (zh) * 2014-03-04 2014-06-04 山东新时代药业有限公司 一种重组甘精胰岛素制剂及其制备方法
AU2015239020A1 (en) 2014-04-04 2016-10-13 Sanofi Substituted indanone compounds as GPR119 modulators for the treatment of diabetes, obesity, dyslipidemia and related disorders
AR099936A1 (es) 2014-04-04 2016-08-31 Sanofi Sa Heterociclos condensados sustituidos como moduladores de gpr119 para el tratamiento de la diabetes, obesidad, dislipidemia y trastornos relacionados
TW201623286A (zh) 2014-04-04 2016-07-01 賽諾菲公司 作為gpr119調控劑用於治療糖尿病、肥胖、血脂異常及相關病症之異吲哚啉酮化合物
PL3169353T3 (pl) 2014-07-16 2020-06-01 Sanofi Biotechnology SPOSOBY LECZENIA PACJENTÓW Z HETEROZYGOTYCZNĄ HIPERCHOLESTEROLEMIĄ RODZINNĄ (heFH)
CN104688678B (zh) * 2015-02-05 2017-11-17 通化东宝药业股份有限公司 一种甘精胰岛素注射液的制备方法及其制备的甘精胰岛素注射液
CN104688677B (zh) * 2015-02-05 2018-02-09 通化东宝药业股份有限公司 一种稳定的甘精胰岛素注射液及其制备方法
WO2016198624A1 (en) 2015-06-12 2016-12-15 Sanofi Exendin-4 derivatives as trigonal glp-1/glucagon/gip receptor agonists
WO2016198628A1 (en) 2015-06-12 2016-12-15 Sanofi Non-acylated exendin-4 derivatives as dual glp-1/glucagon receptor agonists
CA2995645A1 (en) 2015-08-18 2017-02-23 Regeneron Pharmaceuticals, Inc. Anti-pcsk9 inhibitory antibodies for treating patients with hyperlipidemia undergoing lipoprotein apheresis
EA039114B1 (ru) * 2015-11-10 2021-12-06 Санофи-Авентис Дойчланд Гмбх Применение композиции с постоянным соотношением инсулин гларгин/ликсисенатид для лечения сахарного диабета 2 типа
TW201731867A (zh) 2015-12-02 2017-09-16 賽諾菲公司 Fgf21變異體
CN105597087B (zh) * 2016-01-06 2019-04-26 山东新时代药业有限公司 一种甘精胰岛素注射液及其制备方法
GB201607918D0 (en) 2016-05-06 2016-06-22 Arecor Ltd Novel formulations
WO2018055539A1 (en) 2016-09-22 2018-03-29 Wockhardt Limited Pharmaceutical composition containing buffered insulin glargine and glp-1 analogue
AR110299A1 (es) 2016-12-02 2019-03-13 Sanofi Sa Conjugados que comprenden un agonista dual de glp-1 / glucagón, un conector y ácido hialurónico
TW201833132A (zh) 2016-12-02 2018-09-16 法商賽諾菲公司 作為肽類glp1/升糖素/gip三重受體激動劑之新穎化合物
TW201833131A (zh) 2016-12-02 2018-09-16 法商賽諾菲公司 作為胜肽類glp1/升糖素/gip受體促效劑之新化合物
AU2017382038A1 (en) 2016-12-22 2019-08-08 Sanofi FGF21 compound / GLP-1R agonist combinations with optimized activity ratio
US20190031774A1 (en) 2017-06-09 2019-01-31 Sanofi Biotechnology Methods for treating hyperlipidemia in diabetic patients by administering a pcsk9 inhibitor
CN110996905A (zh) * 2017-07-27 2020-04-10 阿道恰公司 至少包含人胰岛素a21g和餐时作用胰高血糖素抑制剂的可注射水溶液形式的组合物
TW202015735A (zh) 2018-05-30 2020-05-01 法商賽諾菲公司 包含glp-1/升糖素/gip三重受體促效劑、連接子及透明質酸之接合物
CN112566655A (zh) 2018-06-21 2021-03-26 赛诺菲 具有优化的活性比率的fgf21化合物/glp-1r激动剂组合
WO2019243628A1 (fr) 2018-06-22 2019-12-26 Adocia Composition injectable a ph 7 comprenant un co-polyaminoacide porteur de charges carboxylates et de radicaux hydrophobes et au moins une insuline basale presentant au moins un effet prandial et un effet basal
CN113993566A (zh) 2019-05-29 2022-01-28 赛诺菲 药物递送装置
IL298415A (en) * 2020-05-22 2023-01-01 Hanmi Pharm Ind Co Ltd Liquid formulations of long-acting conjugate of glucagon, glp-1, and gip trigonal agonist
CA3184546A1 (en) 2020-07-02 2022-01-06 Sanofi Glp-1r agonist / fgf21 fusion proteins
KR20230034356A (ko) 2020-07-02 2023-03-09 사노피 활성이 감소된 glp-1r 효능제 펩티드
CN116209675A (zh) * 2020-08-14 2023-06-02 克云生物科技股份公司 提高生物治疗性分子的功效

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7807242B2 (en) * 2003-12-22 2010-10-05 Novo Nordisk A/S Transparent, flexible, impermeable plastic container for storage of pharmaceutical liquids
US20120122774A1 (en) * 2010-05-19 2012-05-17 Sanofi Long-acting formulations of insulins

Family Cites Families (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4960702A (en) 1985-09-06 1990-10-02 Codon Methods for recovery of tissue plasminogen activator
PH23446A (en) 1986-10-20 1989-08-07 Novo Industri As Peptide preparations
DE3837825A1 (de) * 1988-11-08 1990-05-10 Hoechst Ag Neue insulinderivate, ihre verwendung und eine sie enthaltende pharmazeutische zubereitung
CH682806A5 (de) * 1992-02-21 1993-11-30 Medimpex Ets Injektionsgerät.
CH682805A5 (de) * 1992-02-24 1993-11-30 Medimpex Ets Anzeigeeinrichtung für ein Injektionsgerät.
MX9603936A (es) 1994-03-07 1997-05-31 Inhale Therapeutic Syst Metodos y composiciones para el suministro pulmonar de insulina.
AR002976A1 (es) * 1995-03-31 1998-05-27 Lilly Co Eli Formulaciones farmaceuticas parenterales de efecto prolongado de insulina; cristales de dichos analogos aplicables en dichas formulaciones yprocedimiento de las formulaciones mencionadas
ZA984697B (en) 1997-06-13 1999-12-01 Lilly Co Eli Stable insulin formulations.
CA2306877A1 (en) 1997-10-24 1999-05-06 Eli Lilly And Company Insoluble insulin compositions
US6978471B1 (en) 1999-05-25 2005-12-20 Thomson Licensing S.A. System for acquiring and processing broadcast programs and program guide data
EP1076066A1 (en) 1999-07-12 2001-02-14 Zealand Pharmaceuticals A/S Peptides for lowering blood glucose levels
US6528486B1 (en) 1999-07-12 2003-03-04 Zealand Pharma A/S Peptide agonists of GLP-1 activity
US7022674B2 (en) 1999-12-16 2006-04-04 Eli Lilly And Company Polypeptide compositions with improved stability
DE60134251D1 (de) 2000-09-18 2008-07-10 Sanos Bioscience As Verwendung von glp-2-peptiden
US6852694B2 (en) 2001-02-21 2005-02-08 Medtronic Minimed, Inc. Stabilized insulin formulations
US20030026872A1 (en) 2001-05-11 2003-02-06 The Procter & Gamble Co. Compositions having enhanced aqueous solubility and methods of their preparation
FR2827604B1 (fr) 2001-07-17 2003-09-19 Sanofi Synthelabo Nouveaux derives de 1-phenylsulfonyl-1,3-dihydro-2h-indol-2- one, un procede pour leur preparation et les compositions pharmaceutiques en contenant
WO2003020201A2 (en) * 2001-08-28 2003-03-13 Eli Lilly And Company Pre-mixes of glp-1 and basal insulin
US20100069293A1 (en) 2002-02-27 2010-03-18 Pharmain Corporation Polymeric carrier compositions for delivery of active agents, methods of making and using the same
US7115563B2 (en) 2002-05-29 2006-10-03 Insignion Holding Limited Composition and its therapeutic use
DE10227232A1 (de) * 2002-06-18 2004-01-15 Aventis Pharma Deutschland Gmbh Saure Insulinzubereitungen mit verbesserter Stabilität
CA2490564A1 (en) 2002-07-04 2004-01-15 Zealand Pharma A/S Glp-1 and methods for treating diabetes
DE10235168A1 (de) 2002-08-01 2004-02-12 Aventis Pharma Deutschland Gmbh Verfahren zur Reinigung von Preproinsulin
GB0309154D0 (en) 2003-01-14 2003-05-28 Aventis Pharma Inc Use of insulin glargine to reduce or prevent cardiovascular events in patients being treated for dysglycemia
WO2004064662A2 (en) 2003-01-21 2004-08-05 Ortho-Tain Inc Orthodontic appliance
GB0304822D0 (en) * 2003-03-03 2003-04-09 Dca Internat Ltd Improvements in and relating to a pen-type injector
WO2004078197A1 (en) 2003-03-04 2004-09-16 The Technology Development Company Ltd. Delivery system for drug and cell therapy
JP2007504167A (ja) 2003-08-29 2007-03-01 セントカー・インコーポレーテツド 抗組織因子抗体を用いる移植片の生存を向上する方法
EP1701714A2 (en) 2004-01-07 2006-09-20 Nektar Therapeutics Improved sustained release compositions for pulmonary administration of insulin
WO2005097175A2 (en) 2004-03-31 2005-10-20 Centocor, Inc. Human glp-1 mimetibodies, compositions, methods and uses
AU2005337493A1 (en) 2004-12-22 2007-04-26 Centocor, Inc. GLP-1 agonists, compositions, methods and uses
US7879361B2 (en) 2005-01-04 2011-02-01 Gp Medical, Inc. Nanoparticles for drug delivery
EP1945142B1 (en) 2005-09-26 2013-12-25 Medtronic, Inc. Prosthetic cardiac and venous valves
US8084420B2 (en) 2005-09-29 2011-12-27 Biodel Inc. Rapid acting and long acting insulin combination formulations
US8530447B2 (en) 2005-10-24 2013-09-10 Nestec S.A. Dietary fiber formulation and method of administration
CA2630578C (en) 2005-11-30 2014-04-15 Generex Pharmaceuticals Inc. Orally absorbed pharmaceutical formulation and method of administration
US20100029558A1 (en) 2005-12-06 2010-02-04 Bristow Cynthia L Alpha1 proteinase inhibitor peptides methods and use
CN101454019A (zh) * 2006-04-12 2009-06-10 百达尔公司 速效和长效胰岛素联合制剂
EP2021368A4 (en) 2006-06-08 2010-01-20 Diabecore Medical Inc INSULIN OLIGOMERS DERIVED
DE102006031962A1 (de) * 2006-07-11 2008-01-17 Sanofi-Aventis Deutschland Gmbh Amidiertes Insulin Glargin
US8900555B2 (en) * 2006-07-27 2014-12-02 Nektar Therapeutics Insulin derivative formulations for pulmonary delivery
WO2008124522A2 (en) 2007-04-04 2008-10-16 Biodel, Inc. Amylin formulations
WO2009023566A2 (en) 2007-08-09 2009-02-19 Genzyme Corporation Method of treating autoimmune disease with mesenchymal stem cells
ES2558930T3 (es) 2007-08-13 2016-02-09 Novo Nordisk A/S Análogos de la insulina de acción rápida
JP5241849B2 (ja) * 2007-11-16 2013-07-17 ノボ・ノルデイスク・エー/エス インスリン及びインスリン分泌性ペプチドを含む薬学的組成物
AU2009204309B2 (en) 2008-01-04 2012-11-22 Biodel, Inc. Insulin formulations for insulin release as a function of tissue glucose levels
JP5781308B2 (ja) 2008-02-19 2015-09-16 バイオコン・リミテッドBiocon Limited 異種タンパク質を得る方法およびインスリン類似体
US8329419B2 (en) 2008-05-23 2012-12-11 Amylin Pharmaceuticals, Llc GLP-1 receptor agonist bioassays
TWI451876B (zh) 2008-06-13 2014-09-11 Lilly Co Eli 聚乙二醇化之離脯胰島素化合物
WO2010028055A1 (en) 2008-09-02 2010-03-11 Biodel, Inc. Insulin with a basal release profile
DE102008053048A1 (de) * 2008-10-24 2010-04-29 Sanofi-Aventis Deutschland Gmbh Kombination von einem Insulin und einem GLP-1-Agonisten
ES2650621T3 (es) 2008-10-17 2018-01-19 Sanofi-Aventis Deutschland Gmbh Combinación de una insulina y un agonista de GLP-1
RU2540922C2 (ru) 2008-10-30 2015-02-10 Ново Нордиск А/С Лечение сахарного диабета с использованием инъекций инсулина с частотой менее одного раза в день
EA029759B1 (ru) 2009-02-13 2018-05-31 Бёрингер Ингельхайм Интернациональ Гмбх Антидиабетические лекарственные средства, содержащие ингибитор dpp-4 (линаглиптин) необязательно в комбинации с другими антидиабетическими средствами
US8709400B2 (en) 2009-07-27 2014-04-29 Washington University Inducement of organogenetic tolerance for pancreatic xenotransplant
SG178195A1 (en) 2009-07-31 2012-03-29 Sanofi Aventis Deutschland Long acting insulin composition
TWI468171B (zh) 2009-11-13 2015-01-11 Sanofi Aventis Deutschland 含glp-1激動劑及甲硫胺酸之醫藥組成物
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
DK2329848T4 (da) 2009-11-13 2019-09-09 Sanofi Aventis Deutschland Lixisenatid som supplerende behandling til insulin glargin og metformin til behandling af type 2-diabetes
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
PE20121362A1 (es) 2009-11-13 2012-10-17 Sanofi Aventis Deutschland Composicion farmaceutica que comprende despro36exendina-4(1-39)-lys6-nh2, insulina gly(a21)-arg(b31)-arg(b32) y metionina
WO2011075623A1 (en) 2009-12-18 2011-06-23 Latitude Pharmaceuticals, Inc. One - phase gel compos ition compri s ing phos pholi pids
JP2013520175A (ja) 2010-02-22 2013-06-06 ケース ウェスタン リザーブ ユニバーシティ 可溶形および結晶形の長時間作用型インスリン類似体製剤
AR081066A1 (es) 2010-04-02 2012-06-06 Hanmi Holdings Co Ltd Conjugado de insulina donde se usa un fragmento de inmunoglobulina
TW201141513A (en) 2010-04-14 2011-12-01 Sanofi Aventis Insulin-siRNA conjugates
US8637458B2 (en) 2010-05-12 2014-01-28 Biodel Inc. Insulin with a stable basal release profile
WO2011144674A2 (en) 2010-05-20 2011-11-24 Sanofi-Aventis Deutschland Gmbh PHARMACEUTICAL FORMULATION COMPRISING INSULIN GLARGINE AND SBE4-ß-CYD
EP2389945A1 (en) 2010-05-28 2011-11-30 Sanofi-Aventis Deutschland GmbH Pharmaceutical composition comprising AVE0010 and insulin glargine
WO2011160066A1 (en) 2010-06-17 2011-12-22 Regents Of The University Of Minnesota Production of insulin producing cells
US8532933B2 (en) 2010-06-18 2013-09-10 Roche Diagnostics Operations, Inc. Insulin optimization systems and testing methods with adjusted exit criterion accounting for system noise associated with biomarkers
RU2546520C2 (ru) 2010-08-30 2015-04-10 Санофи-Авентис Дойчланд Гмбх Применение ave0010 для производства лекарственного средства для лечения сахарного диабета 2 типа
CA2815419A1 (en) 2010-10-27 2012-05-03 Novo Nordisk A/S Treating diabetes melitus using insulin injections administered with varying injection intervals
WO2012065996A1 (en) 2010-11-15 2012-05-24 Sanofi-Aventis Deutschland Gmbh PHARMACEUTICAL FORMULATION COMPRISING INSULIN GLARGINE AND MALTOSYL-ß-CYCLODEXTRIN
WO2012066086A1 (en) 2010-11-17 2012-05-24 Sanofi-Aventis Deutschland Gmbh PHARMACEUTICAL FORMULATION COMPRISING INSULIN GLARGINE AND SULFOBUTYL ETHER 7-ß-CYCLODEXTRIN
TW201236692A (en) 2011-02-02 2012-09-16 Sanofi Aventis Deutschland Prevention of hypoglycaemia in diabetes mellitus type 2 patients
PT2683406T (pt) 2011-03-11 2019-07-08 Beth Israel Deaconess Medical Ct Inc Anticorpos anti-cd40 e utilização dos mesmos
WO2013050378A1 (en) 2011-10-04 2013-04-11 Sanofi-Aventis Deutschland Gmbh Glp-1 agonist for use in the treatment of stenosis or/and obstruction in the biliary tract

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7807242B2 (en) * 2003-12-22 2010-10-05 Novo Nordisk A/S Transparent, flexible, impermeable plastic container for storage of pharmaceutical liquids
US20120122774A1 (en) * 2010-05-19 2012-05-17 Sanofi Long-acting formulations of insulins

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Cochran, Elaine, et al., "The Use of U-500 in Patients With Extreme Insulin Resistance", Diabetes Care, Volume 28, Number 5, May 2005 (IDS 2/3/12). *
Cochran, Elaine, et al., "The Use of U-500 in Patients With Extreme Insulin Resistance", Diabetes Care, Volume 28,Number 5, May 2005 (IDS 2/3/12). *
http://diabetes.emedtv.com/lantus/generic-lantus.html *

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11167035B2 (en) 2005-12-28 2021-11-09 Novo Nordisk A/S Insulin compositions and method of making a composition
US10117909B2 (en) 2008-10-17 2018-11-06 Sanofi-Aventis Deutschland Gmbh Combination of an insulin and a GLP-1 agonist
US9526764B2 (en) 2008-10-17 2016-12-27 Sanofi-Aventis Deutschland Gmbh Combination of an insulin and a GLP-1-agonist
US9603904B2 (en) 2008-10-30 2017-03-28 Novo Nordisk A/S Treating diabetes melitus using insulin injections with less than daily injection frequency
US10029011B2 (en) 2009-11-13 2018-07-24 Sanofi-Aventis Deutschland Gmbh Pharmaceutical composition comprising a GLP-1 agonist, an insulin and methionine
US10028910B2 (en) 2009-11-13 2018-07-24 Sanofi-Aventis Deutschland Gmbh Pharmaceutical composition comprising a GLP-1-agonist and methionine
US9707176B2 (en) 2009-11-13 2017-07-18 Sanofi-Aventis Deutschland Gmbh Pharmaceutical composition comprising a GLP-1 agonist and methionine
US9345750B2 (en) 2010-05-19 2016-05-24 Sanofi Long-acting formulations of insulin
US9981013B2 (en) 2010-08-30 2018-05-29 Sanofi-Aventis Deutschland Gmbh Use of AVE0010 for the treatment of diabetes mellitus type 2
US10596229B2 (en) 2010-10-27 2020-03-24 Novo Nordisk A/S Method of treating diabetes mellitus by administration, at specifically defined intervals, of a derivative of a naturally occurring insulin or insulin analogue, the derivative having a prolonged profile of action
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
US9408893B2 (en) 2011-08-29 2016-08-09 Sanofi-Aventis Deutschland Gmbh Pharmaceutical combination for use in glycemic control in diabetes type 2 patients
US9364519B2 (en) 2011-09-01 2016-06-14 Sanofi-Aventis Deutschland Gmbh Pharmaceutical composition for use in the treatment of a neurodegenerative disease
US9987332B2 (en) 2011-09-01 2018-06-05 Sanofi-Aventis Deutschland Gmbh Pharmaceutical composition for use in the treatment of a neurodegenerative disease
US9588130B2 (en) 2012-04-18 2017-03-07 Waters Technologies Corporation Methods for quantifying polypeptides using mass spectrometry
US9964545B2 (en) 2012-04-18 2018-05-08 Waters Technologies Corporation Methods for quantifying polypeptides using mass spectrometry
GB2513805B (en) * 2012-04-18 2020-04-01 Waters Technologies Corp Methods for quantifying polypeptides using mass spectrometry
GB2513805A (en) * 2012-04-18 2014-11-05 Waters Technologies Corp Methods for quantifying polypeptides using mass spectrometry
US10241117B2 (en) 2012-04-18 2019-03-26 Waters Technologies Corporation Methods for quantifying polypeptides using mass spectrometry
WO2013158277A1 (en) * 2012-04-18 2013-10-24 Waters Technologies Corporation Methods for quantifying polypeptides using mass spectrometry
US10758592B2 (en) 2012-10-09 2020-09-01 Sanofi Exendin-4 derivatives as dual GLP1/glucagon agonists
US9745360B2 (en) 2012-12-21 2017-08-29 Sanofi Dual GLP1/GIP or trigonal GLP1/GIP/glucagon agonists
US10253079B2 (en) 2012-12-21 2019-04-09 Sanofi Functionalized Exendin-4 derivatives
US9670261B2 (en) 2012-12-21 2017-06-06 Sanofi Functionalized exendin-4 derivatives
US11191722B2 (en) * 2013-04-03 2021-12-07 Sanofi Treatment of diabetes mellitus by long-acting formulations of insulins
US10092513B2 (en) * 2013-04-03 2018-10-09 Sanofi Treatment of diabetes mellitus by long-acting formulations of insulins
US20160030339A1 (en) * 2013-04-03 2016-02-04 Sanofi Treatment of Diabetes Mellitus by Long-Acting Formulations of Insulins
US20220202711A1 (en) * 2013-04-03 2022-06-30 Sanofi Treatment of Diabetes Mellitus by Long-Acting Formulations of Insulins
US10137172B2 (en) * 2013-04-30 2018-11-27 Novo Nordisk A/S Administration regime
US20160058840A1 (en) * 2013-04-30 2016-03-03 Novo Nordisk A/S Novel Administration Regime
US9789165B2 (en) 2013-12-13 2017-10-17 Sanofi Exendin-4 peptide analogues as dual GLP-1/GIP receptor agonists
US9694053B2 (en) 2013-12-13 2017-07-04 Sanofi Dual GLP-1/glucagon receptor agonists
US9751926B2 (en) 2013-12-13 2017-09-05 Sanofi Dual GLP-1/GIP receptor agonists
US9750788B2 (en) 2013-12-13 2017-09-05 Sanofi Non-acylated exendin-4 peptide analogues
US9775904B2 (en) 2014-04-07 2017-10-03 Sanofi Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
US9771406B2 (en) 2014-04-07 2017-09-26 Sanofi Peptidic dual GLP-1/glucagon receptor agonists derived from exendin-4
US9758561B2 (en) 2014-04-07 2017-09-12 Sanofi Dual GLP-1/glucagon receptor agonists derived from exendin-4
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
WO2016001862A1 (en) 2014-07-04 2016-01-07 Wockhardt Limited Extended release formulations of insulins
US9950039B2 (en) 2014-12-12 2018-04-24 Sanofi-Aventis Deutschland Gmbh Insulin glargine/lixisenatide fixed ratio formulation
US10434147B2 (en) 2015-03-13 2019-10-08 Sanofi-Aventis Deutschland Gmbh Treatment type 2 diabetes mellitus patients
US10159713B2 (en) 2015-03-18 2018-12-25 Sanofi-Aventis Deutschland Gmbh Treatment of type 2 diabetes mellitus patients
US10806797B2 (en) 2015-06-05 2020-10-20 Sanofi Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate
US9982029B2 (en) 2015-07-10 2018-05-29 Sanofi Exendin-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
US10335464B1 (en) 2018-06-26 2019-07-02 Novo Nordisk A/S Device for titrating basal insulin

Also Published As

Publication number Publication date
JOP20150245B1 (ar) 2021-08-17
AU2011202239B2 (en) 2012-06-07
EP3824876A1 (en) 2021-05-26
CN107583038A (zh) 2018-01-16
DOP2012000261A (es) 2013-01-31
US20120122774A1 (en) 2012-05-17
PH12015501421A1 (en) 2015-09-07
MX2012011278A (es) 2012-11-06
US20160228516A1 (en) 2016-08-11
CL2012003197A1 (es) 2013-01-25
TWI605825B (zh) 2017-11-21
KR101913672B1 (ko) 2018-11-01
IL251318A0 (en) 2017-05-29
EP2387989A3 (en) 2012-04-18
PH12015501421B1 (en) 2015-09-07
US20190388511A1 (en) 2019-12-26
EP3824876B1 (en) 2023-02-22
GT201200287A (es) 2015-03-09
US9345750B2 (en) 2016-05-24
ZA201206998B (en) 2013-05-29
BR122019006884B1 (pt) 2022-08-23
PL2387989T3 (pl) 2014-12-31
TN2012000478A1 (en) 2014-04-01
HK1202428A1 (en) 2015-10-02
JP2013147509A (ja) 2013-08-01
TW201607552A (zh) 2016-03-01
JO2944B1 (en) 2016-03-15
CY1115649T1 (el) 2017-01-04
SI2387989T1 (sl) 2014-10-30
BR112012029131B1 (pt) 2020-03-10
DK2571517T3 (en) 2018-07-23
EP2781212B1 (en) 2018-04-04
HUE039355T2 (hu) 2018-12-28
TR201809475T4 (tr) 2018-07-23
JP2018044003A (ja) 2018-03-22
US20140206611A1 (en) 2014-07-24
SI2571517T1 (en) 2018-08-31
CN102319422A (zh) 2012-01-18
WO2011144673A3 (en) 2012-05-03
CN103948913A (zh) 2014-07-30
TWI469787B (zh) 2015-01-21
HK1210435A1 (en) 2016-04-22
RU2015132860A (ru) 2017-02-09
EP3424491A1 (en) 2019-01-09
MX343489B (es) 2016-11-08
EP2781212A1 (en) 2014-09-24
CR20120578A (es) 2013-03-04
AU2011202239C1 (en) 2017-03-16
RS53559B1 (en) 2015-02-27
IL251318B (en) 2018-12-31
AR081200A1 (es) 2012-07-04
EP3607935A1 (en) 2020-02-12
KR20140109515A (ko) 2014-09-15
KR20130040824A (ko) 2013-04-24
CY1120407T1 (el) 2019-07-10
EP4218723A2 (en) 2023-08-02
CY1121042T1 (el) 2019-12-11
US20190046616A1 (en) 2019-02-14
RU2642662C2 (ru) 2018-01-25
MX2019003693A (es) 2019-07-04
PL3824876T3 (pl) 2024-02-19
EP3400931A1 (en) 2018-11-14
DK2781212T3 (en) 2018-07-23
HK1248593A1 (zh) 2018-10-19
LT2781212T (lt) 2018-07-25
US20160339084A1 (en) 2016-11-24
LT2571517T (lt) 2018-07-25
WO2011144673A2 (en) 2011-11-24
CO6640217A2 (es) 2013-03-22
JP5269945B2 (ja) 2013-08-21
CN107583037A (zh) 2018-01-16
NZ602541A (en) 2013-11-29
TW201735941A (zh) 2017-10-16
RU2564104C2 (ru) 2015-09-27
ES2676401T3 (es) 2018-07-19
RU2011119988A (ru) 2012-11-27
JP2015172068A (ja) 2015-10-01
EP2571517B1 (en) 2018-04-04
PL2571517T3 (pl) 2018-09-28
HK1198428A1 (en) 2015-04-24
JP2020040990A (ja) 2020-03-19
UY33391A (es) 2011-10-31
MY156188A (en) 2016-01-29
HUE039354T2 (hu) 2018-12-28
ES2945208T3 (es) 2023-06-29
EP4218723A3 (en) 2023-11-08
SG185593A1 (en) 2012-12-28
CA2792669C (en) 2015-09-29
KR20180088752A (ko) 2018-08-06
TW201208695A (en) 2012-03-01
KR101989343B1 (ko) 2019-06-14
EP2387989B1 (en) 2014-07-16
EP3636250A1 (en) 2020-04-15
DK2387989T3 (da) 2014-10-20
US20210308225A1 (en) 2021-10-07
PT2571517T (pt) 2018-07-10
US20200384087A1 (en) 2020-12-10
HK1248592A1 (zh) 2018-10-19
PL2781212T3 (pl) 2018-08-31
US20200215163A1 (en) 2020-07-09
EP3824876B8 (en) 2023-08-02
EP2387989A2 (en) 2011-11-23
CA2792669A1 (en) 2011-11-24
PH12019500517A1 (en) 2020-03-02
NI201200152A (es) 2013-01-24
BR112012029131A2 (pt) 2016-09-13
JP2011241213A (ja) 2011-12-01
HRP20181028T1 (hr) 2018-08-24
US20150164999A1 (en) 2015-06-18
PE20130511A1 (es) 2013-05-02
KR20170072960A (ko) 2017-06-27
EP2571517A2 (en) 2013-03-27
IL221964A (en) 2017-04-30
TR201809514T4 (tr) 2018-07-23
RU2017146048A3 (ru) 2021-07-01
PE20160781A1 (es) 2016-08-26
KR101486743B1 (ko) 2015-01-28
RU2017146048A (ru) 2019-06-27
JP6333775B2 (ja) 2018-05-30
AU2011202239A1 (en) 2011-12-08
CN104622788A (zh) 2015-05-20
PT2387989E (pt) 2014-09-15
SI2781212T1 (en) 2018-08-31
ES2690302T3 (es) 2018-11-20
TWI496580B (zh) 2015-08-21
HRP20140860T1 (hr) 2014-10-24
ECSP12012298A (es) 2012-12-28
HRP20181026T1 (hr) 2018-08-24
ES2509943T3 (es) 2014-10-20
MA34224B1 (fr) 2013-05-02
TW201427684A (zh) 2014-07-16
SG10201510406YA (en) 2016-01-28
PT2781212T (pt) 2018-07-10

Similar Documents

Publication Publication Date Title
US20210308225A1 (en) Long-Acting Formulations of Insulin
AU2012216648B2 (en) Long-acting formulations of insulins
AU2014203421B2 (en) Long-acting formulations of insulins
OA16421A (en) Long-acting formulations of insulins.

Legal Events

Date Code Title Description
AS Assignment

Owner name: SANOFI, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BECKER, REINHARD;FRICK, ANNKE;BODERKE, PETER;AND OTHERS;SIGNING DATES FROM 20110603 TO 20110621;REEL/FRAME:026785/0991

AS Assignment

Owner name: SANOFI, FRANCE

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE 9TH INVENTOR'S EXECUTION DATE PREVIOUSLY RECORDED ON REEL 026785 FRAME 0991. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNORS:BECKER, REINHARD;FRICK, ANNKE;BODERKE, PETER;AND OTHERS;SIGNING DATES FROM 20110609 TO 20110621;REEL/FRAME:027223/0939

STCB Information on status: application discontinuation

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