WO2011109787A1 - Methods of administering insulinotropic peptides - Google Patents

Methods of administering insulinotropic peptides Download PDF

Info

Publication number
WO2011109787A1
WO2011109787A1 PCT/US2011/027315 US2011027315W WO2011109787A1 WO 2011109787 A1 WO2011109787 A1 WO 2011109787A1 US 2011027315 W US2011027315 W US 2011027315W WO 2011109787 A1 WO2011109787 A1 WO 2011109787A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
concentration
administering
insulinotropic peptide
peptide
Prior art date
Application number
PCT/US2011/027315
Other languages
French (fr)
Inventor
Thomas R. Ulich
Maggie Wang
Jean-Philippe Estradier
Original Assignee
Conjuchem, Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Conjuchem, Llc filed Critical Conjuchem, Llc
Publication of WO2011109787A1 publication Critical patent/WO2011109787A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic 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
    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • 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
    • 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
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin

Landscapes

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

Abstract

The present invention provides methods of administering pharmaceutical formulations comprising an insulinotropic peptide or modified insulinotropic peptide, particularly a conjugate of albumin to exendin-4, or a derivative thereof. The present invention also provides methods or reducing to and maintaining within a target range the blood glucose level of a subject, and for treating diabetes and related diseases or conditions, by administering a pharmaceutical formulation comprising an insulinotropic peptide or modified insulinotropic peptide in accordance with an administration regimen provided herein.

Description

METHODS OF ADMINISTERING INSULINOTROPIC PEPTIDES
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims benefit of U.S. Provisional Patent Application No. 61/311,208, filed March 5, 2010, which is incorporated by reference herein in its entirety.
FIELD OF THE INVENTION
[0002] Methods of administering an insulinotropic peptide or modified insulinotropic peptide are provided. The methods of administering are useful in achieving and maintaining metabolic control in a subject in need thereof, and in the treatment of diabetes and other diseases treatable with an insulinotropic peptide or modified insulinotropic peptide.
BACKGROUND OF THE INVENTION
[0003] The prevalence of diabetes for all age groups worldwide was estimated to be 2.8%, or 171 million in 2000, and is projected to be 4.4%, or 366 million in 2030. See Wild et al, 2004, Diabetes Care 27(5): 1047-1053. In the United States alone, the prevalence of diabetes mellitus in 2005 was estimated at 20.8 million, or roughly 7% of the U.S. population. See Centers for Disease Control and Prevention, 2005, National Diabetes Fact Sheet: General Information and National Estimates on Diabetes in the United States, 2005. Approximately 95% of all subjects with diabetes mellitus have type II disease. Diabetes is currently the fifth leading cause of death in the United States and is associated with excess morbidity stemming from cardiovascular disease, kidney failure, blindness, and lower limb amputation.
[0004] Similarly, obesity is a condition increasingly affecting the population worldwide.
According to the World Health Organization, in 1995 there were an estimated 200 million obese adults worldwide and another 18 million under-five children classified as overweight. As of 2000, the number of obese adults had increased to over 300 million. See Formiguera et al. , 2004, Best Practice & Research Clinical Gastroenterology, 18:6, 1125-1146.
[0005] Incretins and incretin mimetics, such as the insulinotropic peptides GLP-1, exendin-3 and exendin-4, and analogs thereof, have been investigated as possible therapeutic agents for the management of type II non-insulin-dependent diabetes mellitus as well as related metabolic disorders, such as obesity. See, e.g., U.S. Patent No. 6,329,336; and U.S. Patent No. 7,153,825. Recently, it has been shown that conjugation of insulinotropic peptides to albumin can provide longer duration of action in vivo while maintaining their low toxicity and therapeutic advantages. See, e.g., Giannoukakis, Curr Opin Investig Drugs. 4(10): 1245-9 (2003); ConjuChem Press Release, "ConjuChem Reports Final Data Confirming PC-DAC™:Exendin-4 Lowers Glucose and Weight in Phase II Diabetes Trials" (February 9, 2009).
[0006] To date, dosing regimens for insulinotropic peptides have been developed with a titration-to-tolerability fixed-dosing paradigm, with target doses generally established by nausea/vomiting rates observed within a dosed population. For example, dosing of Byetta®, (exenatide) the first incretin mimetic approved in the United States and the European Union, is initiated at 5 micrograms ^g) twice a day, with a possible step up to 10 μg twice a day after 4 weeks. In three Phase III comparative efficacy trials for exenatide, severe nausea was observed in 2% to 5% of the dosed population, with severe nausea generally occurring more frequently in groups that received 10 μg of exantide compared with those who received 5 μg. See, e.g., Iltz et al., Clin Ther. 28(5):652-65 (2006). In another example, dosing of Victoza® (liraglutide), the other incretin mimetic approved in the United States and the European Union, starts at 0.6 milligrams (mg) once daily, which can be increased to 1.2 mg once daily after 1 week and 1.8 mg once daily after 1 week of 1.2 mg dosing. It was previously reported that nausea was experienced by 11%, 16%, and 19% of subjects dosed with 0.6, 1.2 and 1.8 mg of Liraglutide, respectively. See Nauck et al., Diabetes Care 32(1): 84-90 (2009). Other incretins under Phase 2b and Phase 3 clinical development employ administration regimens which consist of 1-3 fixed dose levels.
[0007] Thus, the target dose of a titration-to-tolerance regimen is largely defined by the dose at which an acceptable percentage of patients in the population experience an incidence of gastrointestinal side effect, e.g., a relatively low percentage of patients experience nausea or vomiting. Correspondingly, a significant portion of the population should be able to tolerate higher doses of drug without experiencing adverse events. Subjects having a relatively higher degree of tolerance to the GI effects of the drug can benefit from administration regimens which are not limited by prefixed doses. Such dosing regimens would deliver higher doses of drug over a shorter period time to attain blood glucose levels within a target therapeutic range,
notwithstanding the intolerance of the individual patient to adverse events such as nausea, vomiting, diarrhea, etc.
[0008] Thus, there is a need in the art for individual patient driven treat-to-therapeutic-target dosing regimens for the administration of insulinotropic peptide based therapeutics.
SUMMARY OF THE INVENTION
[0009] Provided herein are methods of treating pre-diabetes and diabetes in a subject. The methods comprise administering to the subject an insulinotropic peptide or modified
insulinotropic peptide. The insulinotropic peptide or modified insulinotropic peptide is administered using a dose-titration-to-target administration regimen which is designed to achieve metabolic control with greater efficacy compared to fixed-interval, fixed-dose, i.e., titration-to- tolerance administration regimens.
[0010] Provided herein methods of treating a human subject having a fasting blood glucose level characteristic of the presence of diabetes, said method comprising increasing the relative amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity, administered to the subject unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject.
In some embodiments, if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
[0011] In one aspect, provided herein is a method of treating a human subject having a fasting blood glucose level characteristic of the presence of diabetes, said method comprising:
(a) determining a value of the fasting blood glucose level of said subject, by a method comprising measuring said fasting blood glucose level, wherein said determining is performed by said subject;
(b) within 24 hours of said determining step, administering to the subject an amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity; wherein said administering is performed by said subject using a flexible dosage device, and wherein said administering is the first administering of said compound that occurs subsequent to said determining step;
(c) after said administering step, repeating steps (a) and (b) for a plurality of cycles, wherein each occurrence of said administering step in a cycle is with said amount being increased relative to the amount of said compound administered in the most recent cycle of said determining and administering steps, unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject; wherein if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject. [0012] In some embodiments, the fasting blood glucose level characteristic of the presence of diabetes is a level of greater than 125 mg/dL. In some embodiments, if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 70 to 125 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
[0013] Provided herein methods of treating a human subject having a fasting blood glucose level characteristic of the presence of pre-diabetes, said method comprising increasing the relative amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity, administered to the subject unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject.
In some embodiments, if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
[0014] In another aspect, provided herein is a method of treating a human subject having a fasting blood glucose level characteristic of the presence of pre-diabetes, said method comprising:
(a) determining a value of the fasting blood glucose level of said subject, by a method comprising measuring said fasting blood glucose level, wherein said determining is performed by said subject;
(b) within 24 hours of said determining step, administering to the subject an amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity; wherein said administering is performed by said subject using a flexible dosage device, and wherein said administering is the first administering of said compound that occurs subsequent to said determining step;
(c) after said administering step, repeating steps (a) and (b) for a plurality of cycles, wherein each occurrence of said administering step in a cycle is with said amount being increased relative to the amount of said compound administered in the most recent cycle of said
determining and administering steps, unless one or more of the following occurs: (i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of pre-diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject; wherein if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
[0015] In some embodiments, the fasting blood glucose level characteristic of the presence of pre-diabetes is a level in the range of 100 mg/dL to 125 mg/dL. In some embodiments, if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 70 to 99 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
[0016] In some embodiments, the modified insulinotropic peptide has an increased half-life relative to the non-modified insulinotropic peptide.
[0017] In some embodiments, if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 80 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject. In some embodiments, if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 75 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject. In some embodiments, if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 70 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
[0018] In some embodiments, if during step (c), the subject experiences a condition of diarrhea or dyspepsia intolerable to the subject, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject. [0019] In some embodiments, the value of the fasting blood glucose level of the subject is the measure of the fasting blood glucose level of said subject obtained in said measuring step. In some embodiments, the value of the fasting blood glucose level of the subject is an average of a plurality of measures of the fasting blood glucose level of said subject over a period of, wherein said plurality of measures comprises said measure obtained in said measuring step.
[0020] In some embodiments, the administering of step (b) is performed within three (3) hours of determining the fasting blood glucose level of the subject in step (a).
[0021] In some embodiments, each cycle of (c) comprises performing said determining and administering steps once every 1, 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, or 13 days. In some embodiments, each cycle of (c) comprises performing said determining and administering steps once a week or once every 2, 3, or 4 weeks, or once a month.
[0022] In some embodiments, the dose of step (b) is at least about 0.5 mg of the insulinotropic peptide or modified insulinotropic peptide. In some embodiments, the dose of step (b) is about
1.5 mg of the insulinotropic peptide or modified insulinotropic peptide.
[0023] In particular embodiments, each cycle of step (c) comprises administering said dose within three (3) hours of determining the fasting blood glucose level of the subject.
[0024] In some embodiments, the repeating of step (c) comprises decreasing said dose relative to a prior dose. In some embodiments, the repeating of step (c) comprises decreasing said dose relative to the most recently administered dose.
[0025] In some embodiments, the repeating of step (c) comprises repeating said determining and administering steps for at least 4, 5, 10 or 20 cycles.
[0026] In some embodiments, each cycle of step (c) comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide which is about 0.1, 0.2, 0.3, 0.4 or 0.5 mg greater than the previously administered dose, e.g., the most recent prior administered dose. In some embodiments, the dose administered during step (c) dose does not exceed about 5.0 mg of the insulinotropic peptide or modified insulinotropic peptide.
[0027] In some embodiments, the insulinotropic peptide is selected from the group consisting of a glucagon-like-peptide 1 (GLP-1) peptide, an exendin peptide, and analogs thereof. In particular embodiments, the exendin peptide is exendin-4 or an analog thereof. In particular embodiments, the modified insulinotropic peptide is a conjugate of albumin and an exendin-4 peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence.
[0028] In some embodiments, administering the modified insulinotropic peptide comprises administering a pharmaceutical formulation comprising: a conjugate of albumin and an exendin-4 peptide, said exendin-4 peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence, said conjugate being at a concentration of about 1 mg/ml to about 100 mg/ml; optionally a buffer; a tonicity modifier, wherein the tonicity modifier is at a concentration of at least 1 mM; a stabilizer; a surfactant, and optionally a preservative, wherein said formulation has a pH from about 4 to about 8. In particular embodiments, the buffer is at a concentration of no more than 0.05 mM, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0. In particular embodiments, the buffer is at a concentration of no more than 0.05 mM, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5. In particular embodiments, the buffer is at a concentration of no more than 0.05 mM, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. In particular embodiments, the buffer is at a concentration of no more than 0.05 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 15 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. In particular embodiments, the buffer is L-histidine at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5. In particular embodiments, the buffer is L-histidine at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. In particular embodiments, the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0. In particular embodiments, the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5. In particular embodiments, the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. In particular embodiments, the buffer is sodium acetate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
[0029] In some embodiments, administering the modified insulinotropic peptide comprises administering a pharmaceutical formulation comprising: a conjugate of albumin and an exendin-4 peptide, said exendin-4 peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence, said conjugate being at a concentration of about 1 mg/ml to about 100 mg/ml; optionally a buffer; a tonicity modifier, wherein the tonicity modifier is at a concentration of at least 1 mM; a stabilizer; a surfactant, and optionally a preservative, wherein said formulation has a pH from about 4 to about 8. In particular embodiments, the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0. In particular embodiments, the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5. In particular embodiments, the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. In particular embodiments, the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 15 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. In particular embodiments, the buffer is L-histidine at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5. In particular embodiments, the buffer is L-histidine at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. In particular embodiments, the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0. In particular embodiments, the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5. In particular embodiments, the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. In particular embodiments, the buffer is sodium acetate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
[0030] In some embodiments, the conjugate comprises albumin cysteine 34 thiol covalently linked to a [2-[2-[2-maleimidopropionamido(ethoxy)ethoxy]acetic acid linker covalently linked to the epsilon amino of a lysine of said exendin-4 peptide. In some embodiments, the lysine has been added to the native exendin-4 sequence. In some embodiments, the lysine has been added to the carboxy terminus of the native exendin-4 sequence.
[0031 In particular embodiments, the conjugate is according to the following:
Figure imgf000010_0001
(SEQ ID NO: 31) wherein X is S, O, or NH of an amino acid of albumin. In some embodiments, X is cysteine 34 thiol of albumin. In some embodiments, the albumin is human serum albumin. In some embodiments, the albumin is recombinant human albumin.
[0032] In some embodiments, the conjugate comprises recombinant human serum albumin cysteine 34 thiol covalently linked to a [2- [2- [2 maleimidopropionamido(ethoxy)ethoxy] acetic acid linker covalently linked to the epsilon amino of the carboxy terminal lysine of exendin-4(l- 39)Lys40-NH2. In some embodiments, conjugate is at a concentration from about 1 mg/ml to about 50 mg/ml. In some embodiments, the conjugate is at a concentration from about 1 mg/ml to about 15 mg/ml. In some embodiments, the conjugate is at a concentration from about 1 mg/ml to about 10 mg/ml. In some embodiments, the conjugate is at a concentration of about 10 mg/ml. In some embodiments, the conjugate is at a concentration of about 20 mg/ml.
[0033] In some embodiments, said administering comprises subcutaneously administering the pharmaceutical formulation to the subject. In some embodiments, said administering comprises administering the pharmaceutical formulation to the subject with a pen-type delivery apparatus. In some embodiments, said pen-type delivery apparatus comprises multiple doses of the pharmaceutical formulation. In some embodiments, said subcutaneously administering is made in the abdomen, thigh, or upper arm of the subject.
[0034] In some embodiments, the subject is a human. In some embodiments, the subject is on a stable dose of one or more anti-diabetic agents prior to, but not during, the administering of an insulinotropic peptide or modified insulinotropic peptide as provided herein. In some
embodiments, the subject is on a stable dose of one or more anti-diabetic agents prior to, and during, the administering of an insulinotropic peptide or modified insulinotropic peptide as provided herein. In some embodiments, the subject is on a stable dose of metformin of > 1000 mg metformin daily for at least 3 months prior to, but not during, the administering of an
insulinotropic peptide or modified insulinotropic peptide as provided herein. In some
embodiments, the subject is on a stable dose of metformin of > 1000 mg metformin daily for at least 3 months prior to, and during, the administering of an insulinotropic peptide or modified insulinotropic peptide as provided herein.
[0035] The invention also encompasses kits comprising pharmaceutical formulations and dosage forms useful for the methods of administering an insulinotropic peptide or modified insulinotropic peptide provided herein.
[0036] In the methods described in this application, preferably the determining (of a value of the fasting blood glucose level of the subject) and/or administering of the insulinotropic peptide or modified insulinotropic peptide to the subject is performed by the subject; most preferably both the determining and administering steps are performed by the subject.
DETAILED DESCRIPTION
/. Definitions
[0037] As used herein, the following terms shall have the following meanings unless otherwise specified:
[0038] As used herein, "about" refers to a value that is no more than 10% above or below the value being modified by the term, unless otherwise indicated. For example, the term "about 20 mg/ml" means a range of from 18 mg/ml to 22 mg/ml. Where "about" is used with respect to a H range, for instance, "about pH 5.0," the pH value is no more than 0.2 above or below the pH being modified by the term. Thus, "about pH 5.0" means a range of from pH 4.8 to 5.2. Similarly, "about pH 6.0" means a range of from pH 5.8 to pH 6.2, and "about pH 7.0" means a range of from pH 6.8 to pH 7.2.
[0039] As used herein, "subject" refers to an animal such as a mammal, including but not limited to, a primate (e.g., human), cow, sheep, goat, horse, dog, cat, rabbit, rat, mouse and the like. In preferred embodiments, the subject is human. In certain embodiments, the subject is a non-human animal, for instance, a non-human animal such as a cow, sheep, goat or horse. The subject can be male or female.
[0040] As used herein, "insulinotropic peptide" refers to a peptide that directly or indirectly causes a glucose dependent increase in the amount of insulin release, such that the amount of insulin released from the pancreas is greater when plasma glucose levels are elevated as compared to when plasma glucose levels are normal. Examples of insulinotropic peptides include, but are not limited to, Glucagon-Like Peptide- 1 (GLP-1), exendin-3 and exendin-4, along with their analogs and derivatives.
[0041] As used herein, "modified insulinotropic peptide" refers to an insulinotropic peptide compound modified, e.g., by attachment to one or more non-peptide moieties. Preferably, the modified insulinotropic peptide is modified to have an increased half-life relative to the non- modified insulinotropic peptide. Any method known in the art for increasing the half-life, e.g., the in-vivo half life of a peptide can be used to generate such a modified insulinotropic peptide. In some embodiments, a modified insulinotropic peptide is modified to have an increased half-life relative to the non-modified insulinotropic peptide by conjugating the insulinotropic peptide to albumin. In some embodiments, a modified insulinotropic peptide is modified to have an increased half-life relative to the non-modified insulinotropic peptide by formulating the insulinotropic peptide in a sustained-release or long-acting-release (LAR) pharmaceutical formulation. For example, the insulinotropic peptide may be formulated into microspheres composed of the insulinotropic peptide and a poly(lactide-coglycolide) polymeric matrix.
Poly(lactide-coglycolide) is a common biodegradable medical polymer commonly used in absorbable sutures and extended-release pharmaceuticals. After injection, the active compound, e.g., the insulinotropic peptide, is slowly released from the microspheres through diffusion and erosion.
[0042] As used herein, "insulinotropic" means having insulinotropic activity, i.e., the ability to stimulate, or to cause the stimulation of, the synthesis or expression of the hormone insulin.
Insulinotropic peptides include, but are not limited to, GLP-1, exendin-3, exendin-4, and precursors, derivatives, or fragments of peptides such as GLP-1, exendin-3 and exendin-4 and other peptides with insulinotropic activity.
[0043] "Glucagon-Like Peptide-1" ("GLP-1") and "GLP-1 derivatives" are intestinal hormones and derivatives thereof, respectively, that generally simulate insulin secretion during hyperglycemia, suppress glucagon secretion, stimulate (pro) insulin biosynthesis and decelerate gastric emptying and acid secretion. In some embodiments, the glucagon-like peptide is GLP-1 (7- 37). In some embodiments, the glucagon-like peptide is GLP-1 (7-36). Some GLP-1 peptides and GLP-1 derivatives, such as those described herein as SEQ ID NOS: 3-15, promote glucose uptake by cells but do not simulate insulin expression, as disclosed in U.S. Pat. No. 5,574,008, which is incorporated by reference herein in its entirety.
[0044] "Exendin-3" is a naturally occurring GLP-1 agonist that can be isolated from salivary secretions of Heloderma horridum, the Mexican bearded lizard, and shares a 53% overlap with mammalian GLP-1 amino acid sequence, as disclosed in U.S. Pat. No. 5,424,286, which is incorporated by reference herein in its entirety. The amino acid sequence of exendin-3 is
HSDGTFTSDLSKQMEEEAVRLFIEWLKNGG PSSGAPPPS (SEQ ID NO: 16). The term "exendin-3" as used herein can refer either to exendin-3 purified from a natural source, or it can refer to chemically synthesized exendin-3, or exendin-3 produced by recombinant techniques.
[0045] "Exendin-4" is a naturally occurring GLP-1 agonist that can be isolated from salivary gland venom of Heloderma suspectum, the Gila monster, and shares a 53% overlap with mammalian GLP-1 amino acid sequence as disclosed in U.S. Pat. No. 5,424,286, which is incorporated by reference herein in its entirety. The amino acid sequence of exendin-4 is
HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPS (SEQ ID NO: 17). Exendin-4 decreases glucagons and/or increases insulin secretion in a glucose-dependent manner, and mimics certain actions of GLP-1, including binding to and activating the human GLP-1 receptor. Exendin-4 improves glycemic control by reducing fasting and postprandial glucose
concentrations through restoration of first-phase insulin response, delaying gastric emptying, and decreasing food intake. The term "exendin-4" as used herein can refer either to exendin-4 purified from a natural source, or it can refer to chemically synthesized exendin-4, or exendin-4 produced by recombinant techniques.
[0046] "Reactive groups" are chemical groups capable of forming a covalent bond. Such reactive agents can be coupled or bonded to an insulinotropic peptide of interest to form a modified insulinotropic peptide. Reactive groups can generally be carboxy, phosphoryl, or acyl groups, either as an ester or a mixed anhydride, or an imidate, thereby capable of forming a covalent bond with functionalities such as an amino group, a hydroxy or a thiol at the target site on albumin. For the most part, the esters will include phenolic compounds, or be thiol esters, alkyl esters, phosphate esters, or the like. Reactive groups include succinimidyl and maleimido groups.
[0047] "Functionalities" are groups on albumin with which reactive groups on modified insulinotropic peptides are capable of reacting to form covalent bonds. Functionalities include hydroxyl groups for bonding to ester reactive entities; thiol groups for bonding to maleimides and maleimido groups, imidates and thioester groups; and amino groups for bonding to carboxy, phosphoryl or acyl groups on reactive entities.
[0048] "Linking groups" are chemical moieties that can be used to connect reactive groups to insulinotropic peptides. Linking groups can comprise one or more alkyl groups such as methyl, ethyl, propyl, butyl, etc. groups, alkoxy groups, alkenyl groups, alkynyl groups or amino group substituted by alkyl groups, cycloalkyl groups, polycyclic groups, aryl groups, polyaryl groups, substituted aryl groups, heterocyclic groups, and substituted heterocyclic groups. Linking groups can also comprise poly ethoxy aminoacids such as AEA ((2-amino) ethoxy acetic acid) or a preferred linking group AEEA ([2-(2-amino)ethoxy)]ethoxy acetic acid).
[0049] As used herein, "albumin" refers to the most abundant protein in blood plasma having a molecular weight of approximately between 65 and 67 kilodaltons in its monomeric form, depending on the species of origin. The term "albumin" is used interchangeably with "serum albumin" and is not meant to define the source of the albumin which forms a conjugate with the insulinotropic peptides of the invention. Thus, the term "albumin" as used herein can refer either to albumin purified from a natural source such as blood or serous fluids, or it can refer to chemically synthesized albumin, or albumin produced by recombinant techniques.
[0050] An "insulinotropic peptide conjugate" comprises an insulinotropic peptide that has been conjugated to a protein such as albumin, transferrin, immunoglobulin, immunoglobulin Fc fragment, human chorionic gonadotropin, pseudo-PEG unstructured polypeptide, and elastin-like peptide, etc., or a polymer such as poly-ethylene glycol or other polymer via a covalent bond formed between the insulinotropic peptide and a functionality on the protein or polymer, respectively. In some embodiments, the insulinotropic peptide is coupled to the protein or polymer via a linking group. In some embodiments, the insulinotropic peptide has been modified to contain a reactive group to which a protein or polymer is covalently bound. In some embodiments, the reactive group is coupled to the insulinotropic peptide via a linking group.
[0051] An "insulinotropic peptide albumin conjugate" comprises an insulinotropic peptide that has been conjugated to albumin via a covalent bond formed between the insulinotropic peptide and a functionality on albumin. In some embodiments, the insulinotropic peptide has been modified to contain a reactive group to which albumin is covalently bound. In some
embodiments, the reactive group is coupled to the insulinotropic peptide via a linking group. [0052] "Stable" formulations include formulations in which the insulinotropic peptide or modified insulinotropic peptide, e.g., peptide or peptide conjugate, therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage. Various analytical techniques for measuring protein stability are available in the art and are reviewed in Lee, V., 1991, Peptide and Protein Drug Delivery, 247-301 (Marcel Dekker, Inc., New York, N.Y.) and Jones, A. 1993, Adv. Drug Delivery Rev. 10: 29-90, for example. Stability can be measured at a selected temperature for a selected time period. Preferably, the formulation is stable at room temperature (about 25 °C) or at 40 °C for at least 1, 2, 3, 4, 5, 6, 9, 12, 24 or 36 months and/or stable at about 2-8 °C for at least 1, 2, 3, 4, 5, 6, 9, 12, 24 or 36 months.
Furthermore, in certain embodiments, the formulation is preferably stable following freezing (e.g., -70 °C ). In certain embodiments, the criteria for stability are as follows: (1) the formulation remains clear by visual analysis; (2) the concentration, pH and osmolality of the formulation has no more than about ± 10% change; (3) no more than about 10%, more preferably no more than about 5%, or most preferably no more than about 1% of aggregate forms as measured by SEC- HPLC; and (4) no more than 10%, more preferably no more than about 5%, or most preferably no more than 1% of peptide or peptide conjugate breaks down as measured by SDS-PAGE or RP- HPLC.
[0053] A peptide or peptide conjugate "retains its physical stability" in a pharmaceutical formulation if it shows substantially no signs of aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography. For example, the peptide of a peptide-conjugate retains its physical stability in a pharmaceutical formulation where less than about 10%, more preferably less than about 5, or most preferably less than about 1% of the peptide or peptide conjugate is present as an aggregate in the formulation.
[0054] A peptide or peptide conjugate "retains its chemical stability" in a pharmaceutical formulation if the chemical stability at a given time is such that the peptide is considered to retain its biological activity as defined below. Chemical stability can be assessed by detecting and quantifying chemically altered forms of the peptide. Chemical alteration may involve size modification (e.g. clipping) which can be evaluated using size exclusion chromatography, SDS- PAGE and/or matrix-assisted laser desorption ionization/time-of-flight mass spectrometry (MALDI/TOF MS), for example. Other types of chemical alteration include charge alteration (e.g. occurring as a result of deamidation) which can be evaluated by ion-exchange
chromatography, for example.
[0055] A peptide or peptide conjugate "retains its biological activity" in a pharmaceutical formulation, if the peptide in a pharmaceutical formulation is biologically active for its intended purpose. For example, biological activity is retained if the biological activity of the peptide in the pharmaceutical formulation is at least about 70%, at least about 80%, or more preferably, at least about 90% (within the errors of the assay) of the biological activity exhibited at the time the pharmaceutical formulation was prepared. The biological activity for a particular peptide will be the biological activity of the peptide known to those of skill in the art. For example, the biological activity of GLP-1 includes, but is not limited to, stimulation of insulin secretion during hyperglycemia, suppression of glucagon secretion, stimulation of (pro) insulin biosynthesis, deceleration of gastric emptying and acid secretion, and reduction of blood glucose levels.
[0056] As used herein, a "buffer" refers to a substance that resists changes in pH and maintains the pH value of a solution in an acceptable range by the action of its acid-base conjugate components. Thus, a buffered formulation described herein has a pH in the range from about 4 to about 8; preferably from about 5 to about 7; and most preferably has a pH in the range from about 5 to about 6. In some embodiments, the pH of the buffered formulation is about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9 or 8.0. Examples of buffers that will control the pH in this range include acetate (e.g. sodium acetate), phosphate (e.g. sodium phosphate), succinate (such as sodium succinate), maleate, gluconate, histidine (e.g., L-histidine), citrate, other organic acid buffers, and mixtures thereof, e.g., citrate-phosphate.
[0057] As used herein, a "tonicity modifier" refers to a compound which, in appropriate amount, renders the formulation isotonic, such as, for example, sodium chloride, calcium chloride, magnesium chloride, lactose, sorbitol, sucrose, mannitol, trehalose, raffinose, polyethylene glycol, hydroxyethyl starch, glycine and the like. "Isotonic" is meant that the formulation of interest has essentially the same osmolarity as human blood. Isotonic formulations will generally have an osmolarity from about 250 to 350 mOsm, preferably from about 250 to about 330 mOsm. Osmolarity can be measured using a vapor pressure or ice-freezing type osmometer, for example.
[0058] As used herein, a "surfactant" refers to a compound that reduces interfacial tension between a liquid and a solid when dissolved in solution, which can be added to the formulation to reduce aggregation of the reconstituted protein and/or reduce the formation of particulates in the reconstituted formulation. Examples of surfactants useful for the methods and formulations described herein include polysorbates (e.g. polysorbates 20 or 80); poloxamers (e.g. poloxamer 188 (pluronic F68)); Triton; sodium dodecyl sulfate (SDS); sodium laurel sulfate; sodium octyl glycoside; lauryl-, myristyl-, linoleyl-, or stearylsulfobetaine; lauryl-, myristyl-, linoleyl- or stearyl-sarcosine; linoleyl-, myristyl-, or cetylbetaine; lauroamidopropyl-, cocamidopropyl-, linoleamidopropyl-, myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-betaine (e.g. lauroamidopropyl); myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-dimethylamine; sodium methyl cocoyl-, or disodium methyl oleyl-taurate; and the MONAQUAT™ series (Mona Industries, Inc., Paterson, N.J.), polyethyl glycol, polypropyl glycol, and copolymers of ethylene and propylene glycol, etc.
[0059] As used herein, a "stabilizer" refers to a compound that stabilizes a peptide or peptide conjugate in a pharmaceutical formulation during fluctuations in storage temperature and minimizes degradation products, peptide degradants and aggregation. Useful stabilizers in the formulations described herein include, but are not limited to, sodium octanoate, myristic acid, Na- N-acetyltryptophan, L-glutamic acid, arginine, nitrogen and combinations thereof. In particular embodiments, the stabilizer is sodium octanoate. In particular embodiments, the stabilizer is myristic acid.
[0060] As used herein, a "preservative" refers to a compound which can be added to the formulation to essentially reduce bacterial activity therein, thus facilitating the production of a multi-use formulation, for example. Examples of useful preservatives include m-cresol, benzyl alcohol, methanol, ethanol, iso-propanol, butyl paraben, ethyl paraben, methyl paraben, phenol, glycerol, xylitol, resorcinol, cathechol, 2, 6-dimethylcyclohexanol, 2-methyl-2,4-pentadiol, dextran, polyvinylpyrrolidone, 2-chlorophenol, benzethonium chloride, merthiolate (thimersosal), benzoic acid (propyl paraben) MW 180.2, benzoic acid MW 122.12, benzalkonium chloride, chlorobutanol, sodium benzoate, sodium propionate, and cetylpyridinium chloride.
[0061] As used herein, a "bulking agent" refers to a compound which can add mass to a lyophilized mixture and contributes to the physical structure of a lyophilized cake (e.g. facilitates the production of an essentially uniform lyophilized cake which maintains an open pore structure). Exemplary bulking agents include mannitol, glycine, polyethylene glycol and xorbitol. In addition to providing a pharmaceutically acceptable cake, bulking agents also typically impart useful qualities to the lyophilized composition such as modifying the collapse temperature, providing freeze-thaw protection, further enhancing the protein stability over long-term storage, and the like. These agents can also serve as tonicity modifiers.
[0062] As used herein, a "reducing sugar" is one which contains a hemiacetal group that can reduce metal ions or react covalently with lysine and other amino groups in proteins and a "non- reducing sugar" is one which does not have these properties of a reducing sugar. Examples of reducing sugars are fructose, mannose, maltose, lactose, arabinose, xylose, ribose, rhamnose, galactose and glucose. Nonreducing sugars include sucrose, trehalose, sorbose, melezitose and raffinose. Preferably, lyophilized pharmaceutical formulations as described herein are lyophilized in the absence of reducing sugars, or in the presence of only non-reducing sugars. [0063] As used herein, a "pharmaceutically acceptable carrier" refers to a pharmaceutically acceptable material, composition or vehicle, suitable for administration to mammals, preferably humans. The carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not overly injurious (e.g., fatal) to the subject. In a preferred embodiment, the pharmaceutically acceptable carrier is approved for administration to humans by a government regulatory agency such as the Food and Drug Administration (FDA) or the European Medicines Agency (EMEA).
[0064] "Preventing" or "prevention" of any disease or disorder refers to a reduction in the risk of acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed or predisposed to the disease but does not yet experience or display symptoms of the disease). Preferably, prevention refers to the use of a compound or composition in a subject not yet affected by the disease or disorder or not yet exhibiting a symptom of the disease or disorder, for instance a subject not yet diabetic or not yet exhibiting the symptoms of diabetes.
[0065] "Treating" or "treatment" of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof) that exists in a subject. In another embodiment, "treating" or "treatment" refers to ameliorating at least one physical parameter of the disease or disorder, which may be indiscernible by the subject. In yet another embodiment, "treating or treatment" refers to beneficially modulating the disease, either physically (e.g., stabilization of a discernable symptom) or physiologically (e.g., stabilization of a physical parameter) or both.
[0066] As used herein, an "effective amount," with respect to treatment, means an amount of an insulinotropic peptide or modified insulinotropic peptide, e.g., insulinotropic peptide conjugate, that when administered to a subject for treating a disease is sufficient to treat the disease. An effective amount can vary depending on, inter alia, the insulinotropic compound used, the disease and its severity and the age, weight, etc., of the subject to be treated.
II. Administration Regimens
[0067] In one aspect, provided herein is a method of treating a human subject having a fasting blood glucose level characteristic of the presence of diabetes, said method comprising:
(a) determining a value of the fasting blood glucose level of said subject, by a method comprising measuring said fasting blood glucose level, wherein said determining is performed by said subject; (b) within 24 hours of said determining step, administering to the subject an amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity; wherein said administering is performed by said subject using a flexible dosage device, and wherein said administering is the first administering of said compound that occurs subsequent to said determining step;
(c) after said administering step, repeating steps (a) and (b) for a plurality of cycles, wherein each occurrence of said administering step in a cycle is with said amount being increased relative to the amount of said compound administered in the most recent cycle of said determining and administering steps, unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject;
(iv) wherein if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
[0068] In some embodiments, the fasting blood glucose level characteristic of the presence of diabetes is any fasting blood glucose level known in the art to be indicative of the presence of diabetes. For example, in some embodiments, the fasting blood glucose level characteristic of diabetes is provided by the American Diabetes Association, and is in accordance with the Standards of Medical Care in Diabetes as published annually. See, e.g., "Standards of Medical Care in Diabetes - 2010," Diabetes Care 2010 Jan; 33 Suppl. 1 :S11-61, the entirety of which is incorporated by reference herein. In some embodiments, the fasting blood glucose level characteristic of the presence of diabetes is a level of greater than 125 mg/dL. In some embodiments, the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of diabetes when the fasting blood glucose level of the subject is 125 mg/dL or lower, e.g., about 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 or 125 mg/dL. In some embodiments of the methods provided herein, the fasting blood glucose level of the subject is determined to be below said fasting blood glucose level characteristic of the presence of diabetes when two consecutive determinations of the subject's blood glucose level indicates a fasting blood glucose level of 125 mg/dL or lower, e.g., about 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 or 125 mg/dL. In other embodiments of the methods provided herein, the fasting blood glucose level of the subject is determined to be below said fasting blood glucose level characteristic of the presence of diabetes when the average weekly fasting blood glucose level of the subject is 125 mg/dL or lower, e.g., about 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 or 125 mg/dL.
[0069] In another aspect, provided herein is a method of treating a human subject having a fasting blood glucose level characteristic of the presence of pre-diabetes, said method comprising:
(a) determining a value of the fasting blood glucose level of said subject, by a method comprising measuring said fasting blood glucose level, wherein said determining is performed by said subject;
(b) within 24 hours of said determining step, administering to the subject an amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity; wherein said administering is performed by said subject using a flexible dosage device, and wherein said administering is the first administering of said compound that occurs subsequent to said determining step;
(c) after said administering step, repeating steps (a) and (b) for a plurality of cycles, wherein each occurrence of said administering step in a cycle is with said amount being increased relative to the amount of said compound administered in the most recent cycle of said
determining and administering steps, unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of pre-diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject; wherein if the subject experiences one or more of said (i)— (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
[0070] In some embodiments, the fasting blood glucose level characteristic of the presence of pre-diabetes is any fasting blood glucose level known in the art to be indicative of the presence of pre-diabetes. For example, in some embodiments, the fasting blood glucose level characteristic of pre-diabetes is provided by the American Diabetes Association, and is in accordance with the Standards of Medical Care in Diabetes as published annually. See, e.g., "Standards of Medical Care in Diabetes— 2010," Diabetes Care 2010 Jan; 33 Suppl. 1 :S11-61, the entirety of which is incorporated by reference herein. In some embodiments, the fasting blood glucose level characteristic of the presence of pre-diabetes is a level in the range of 100 mg/dL to 125 mg/dL. In some embodiments, the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of pre-diabetes when the fasting blood glucose level of the subject is below 100 mg/dL, e.g., about 70, 75, 80, 85, 90, 95, 96, 97, 98 or 99 mg/dL. In some embodiments of the methods provided herein, the fasting blood glucose level of the subject is determined to be below said fasting blood glucose level characteristic of the presence of pre- diabetes when two consecutive determinations of the subject's blood glucose level indicates a fasting blood glucose level of below 100 mg/dL, e.g., about 70, 75, 80, 85, 90, 95, 96, 97, 98 or 99 mg/dL. In other embodiments of the methods provided herein, the fasting blood glucose level of the subject is determined to be below said fasting blood glucose level characteristic of the presence of prediabetes when the average weekly fasting blood glucose level of the subject is below 100 mg/dL, e.g., about 70, 75, 80, 85, 90, 95, 96, 97, 98 or 99 mg/dL.
[0071] The amount of an insulinotropic peptide or modified insulinotropic peptide
administered over the course of the administration regimen that will be effective in treating prediabetes or diabetes will vary with the nature and severity of the disorder or condition underlying the high glucose levels of the subject, and the route by which the active ingredient is administered over the course of the administration regimens provided herein. The frequency and dosage will also vary according to the judgment of the practitioner of skill based on factors specific for each subject depending on the age, body weight, response, and the past medical history of the subject.
[0072] The insulinotropic peptide or modified insulinotropic peptide can be administered according to any technique deemed suitable by one of skill in the art. For example, the insulinotropic peptide or modified insulinotropic peptide can be administered by any of the following means: (a) enterally, e.g., orally (by mouth), rectally (e.g., in the form of a suppository or an enema), by feeding tube (e.g., gastric feeding tube, duodenal feeding tube, gastrostromy);
(b) parenterally, e.g., subcutaneously, intravenously, intramuscularly, intradermally (into the skin itself), transdermally (diffusion through skin, e.g., intact skin), intra-arterially, intra-peritoneally, intracardiac ally (into the heart) administration, intraosseously (into the bone marrow) administration intrathecally (into the spinal canal), transmucosally (diffusion through a mucous membrane, e.g., insufflation (snorting), nasally, e.g., intranasally), sublingually (under the tongue), buccally (through the cheek), vaginally, by inhalation (e.g., pulmonary administration);
(c) topically; (d) epidurally (injection or infusion into the epidural space); and (e) intravitreally. Each administration of insulinotropic peptide or modified insulinotropic peptide can be by bolus or by infusion. In preferred embodiments, the insulinotropic peptide or modified insulinotropic peptide is administered subcutaneously. In more preferred embodiments, the insulinotropic peptide or modified insulinotropic peptide is administered subcutaneously in the abdomen, thigh, or upper arm. In some embodiments, the insulinotropic peptide or modified insulinotropic peptide is administered to the subject by the subject. In other embodiments, the insulinotropic peptide or modified insulinotropic peptide is administered to the subject by a physician.
[0073] In a particular embodiment, the insulinotropic peptide or modified insulinotropic peptide is administered subcutaneously using a needle, e.g., a 23-gauge needle, a 24-gauge needle, a 25-gauge needle, a 26-gauge needle, a 27-gauge needle, a 28-gauge needle, a 29-gauge needle, a 30-gauge needle, a 31 -gauge needle, a 32-gauge needle, or a 33-gauge needle, or a higher gauge needle.
[0074] In certain embodiments, the insulinotropic peptide or modified insulinotropic peptide is provided in a liquid formulation in a delivery device for convenient administration of a dose to the subject. Any delivery device known in the art can be used. In certain embodiments, the insulinotropic peptide or modified insulinotropic peptide is provided as a lyophilized formulation and reconstituted prior to administration. In particular embodiments, the lyophilized
insulinotropic peptide or modified insulinotropic peptide is provided in a 2-chamber syringe and reconstituted in the syringe. In particular embodiments, the lyophilized insulinotropic peptide or modified insulinotropic peptide is provided in a 2-chamber cartridge and reconstituted in the cartridge. In particular embodiments, the delivery device is a syringe configured for subcutaneous delivery, e.g. a 0.3, 0.5, 1, 2, 3 or greater than 3 ml syringe having a 23, 25, 25, 26, 27, 28, 29, 30, 31, 32, 33, or larger than 33-gauge needle. In a particular embodiment, the delivery device is a pen-type delivery apparatus comprising a single dose or multiple doses, i.e., is preloaded for use for a single administration or multiple administration events. Such a pen-type delivery apparatus can be permanent, e.g., a permanent pen that houses a disposable cartridge containing a single dose or multiple doses, or the entire apparatus can be disposable, e.g., a disposable pen that contains a single dose or multiple doses. In certain embodiments where the pen-type delivery apparatus comprises multiple doses, the dose can be pre-set, i.e., fixed. In other embodiments, the dose can be a flexible dose, i.e., dialed-in by the user. In some embodiments, the pen-type delivery apparatus comprises a luer-lock, luer-cone, or other needle fitting connector that facilitates attachment of a disposable needle. In other embodiments, the pen-type delivery apparatus comprises a staked, i.e., permanent needle. In another particular embodiment, the container is a syringe. In some embodiments, the syringe comprises a luer-lock, luer-cone, or other needle fitting connector that facilitates attachment of a disposable needle. In other embodiments, the syringe comprises a staked, i.e., permanent, needle. In some embodiments, the syringe is prefilled with a single dose or multiple doses.
A. Determination of a Value of Blood Glucose Level [0075] In some embodiments, step (a) of the methods described herein comprises determining a value of the fasting blood glucose level of the subject, based on a measure of the fasting blood glucose level, comprising measuring the fasting blood glucose level. In some embodiments, the fasting blood glucose levels of the subject may be measured by glucometer readings, e.g., finger stick glucometer readings. In preferred embodiments, the determination of the subject's fasting blood glucose level is performed by the subject. In other embodiments, the determination of the subject's fasting blood glucose level is performed by a physician.
[0076] In some embodiments, the determined value of the fasting blood glucose level of the subject is the measure of the fasting blood glucose level of the subject obtained from a single measuring of said fasting blood glucose level. In other embodiments, the determined value of the fasting blood glucose level of the subject is the average measure of the fasting blood glucose level of the subject obtained from a plurality of measures of the fasting blood glucose level of the subject over a period of time. For example, the determined value of the fasting blood glucose level of the subject can be the average weekly fasting blood glucose level, calculated from the daily fasting blood glucose levels of the subject over a period of seven days. In other
embodiments, the determined value of the fasting blood glucose level of the subject can be the average fasting blood glucose level calculated from the daily fasting blood glucose levels of the subject over a period of 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more days.
[0077] Although the administration regimens described herein provide for a reduction to and maintenance within a target glucose range as indicated by the fasting blood glucose reading(s) of the subject, other indicia of metabolic control can be utilized within the scope of the methods. For example, improvement of the subject's metabolic control can be determined through
measurement of efficacy variables known to those of skill in the art, including, but not limited to glycosylated hemoglobin (HbAic), fasting plasma glucose, ten point blood glucose profiles and 24 hour glucose profiles.
[0078] In some embodiments of the methods provided herein, improvement of the subject's metabolic control is determined by the percentage of glycosylated hemoglobin (FIbAic) of the subject. The formation of glycosylated hemoglobins is non-enzymatic and occurs over the lifespan of a blood cell, which is about 120 days under normal conditions. The amount of FIbAlc is proportional to the concentration of glucose in the blood, and is therefore related to time- averaged glucose concentration over the period prior to the measurement, which is approximately two to three months. FIbAlc values may be used to assess metabolic control, in which short-term fluctuations in blood or plasma glucose levels do not affect the measurement. Thus, in some embodiments, a target glucose range as indicated by FIbAic level is less than 6.5%, and less than 6% in a non-diabetic subject. Measurement of glycosylated hemoglobins may augment other traditional methods of assessing control of glucose metabolism. For example, measurement of glycosylated hemoglobins may be used when urine glucose records are inadequate, when blood glucose levels vary markedly throughout the day or from day to day, and for a new patient with known or suspected diabetes in whom there is no previous record of blood glucose concentration. Methods known in the art for determining levels of glycosylated hemoglobins, include, but are not limited to, ion exchange chromatography, high-performance liquid chromatography, affinity chromatography, colorimetry, radioimmunoassay, electrophoresis, and isoelectric focusing. B. Administration of Starting Dose
[0079] In some embodiments, step (b) of the methods provided herein comprises administering to the subject a starting dose of a pharmaceutical formulation comprising an insulinotropic peptide or modified insulinotropic peptide. The amount of the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be determined by one skilled in the art. Different starting amounts of the insulinotropic peptide or modified insulinotropic peptide may be applicable for different disorders and conditions, or different subjects, as will be readily known by those of ordinary skill in the art. Exemplary doses of an insulinotropic peptide or modified insulinotropic peptide include milligram or microgram amounts of compound per kilogram of subject or sample weight (e.g., about 1 microgram per kilogram to about 50 microgram per kilogram, e.g., about 10 microgram per kilogram to about 30 microgram per kilogram).
[0080] In some embodiments, administration of the starting dose of the insulinotropic peptide or modified insulinotropic peptide is performed within 24 hours of determining the fasting blood glucose level of the subject in step (a) of the methods provided herein. In some embodiments, administration of the starting dose of the insulinotropic peptide or modified insulinotropic peptide is performed within about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours of determining the fasting blood glucose level of the subject. In a particular embodiment, administration of the starting dose of the insulinotropic peptide or modified insulinotropic peptide is performed within about 3 hours of determining the fasting blood glucose level of the subject.
[0081] In some embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide comprises the insulinotropic peptide or modified insulinotropic peptide in an amount between about 500 μg and 3000 μg (e.g., about 525 μg, 550 μg, 575 μg, 600 μg, 625 μg, 650 μg, 675 μg, 700 μg, 725 μg, 750 μg, 775 μg, 800 μg, 825 μg, 850 μg, 875 μg, 900 μg, 925 μg, 950 μg, 975 μg, 1000 μg, 1025 μg, 1050 μg, 1075 μg, 1100 μg, 1125 μg, 1150 μg, 1175 μg, 1200 μg, 1225 μg, 1250 μg, 1275 μg, 1300 μg, 1325 μg, 1350 μg, 1375 μg, 1400 μg, 1425 μg, 1450 μg, 1475 μg, 1500 μg, 1525 μg, 1550 μg, 1575 μg, 1600 μg , 1625 μg, 1650 μg, 1675 μg, 1700 μg, 1725 μg, 1750 μg, 1775 μg, 1800 μg, 1825 μg, 1850 1875 μ , 1900 μg, 1925 μ^ 1950 μg, 1975 μ^ 2000 μ , 2025 μg, 2050 μ^ 2075 μg, 2100 μ^ 2125 μ , 2150 μg, 2175 μ§, 2200 μg, 2225 μ , 2250 μ , 2275 μg, 2300 μ , 2325 μg, 2350 μ , 2375 μ , 2400 μg, 2425 μg, 2450 μg, 2475 μg, 2500 μg, 2525 μg, 2550 μg, 2575 μg, 2600 μg, 2625 μg, 2650 μg, 2675 μg, 2700 μ^ 2725 μg, 2750 μg, 2775 μ^ 2800 μg, 2825 μg, 2850 μg, 2875 μg, 2900 μg, 2925 μg, 2950 μg, or 2975 μg), preferably between about 1000 μg and 2750 μg (e.g., about 1025 μg, 1050 μg, 1075 μg, 1100 μg, 1125 μg, 1150 μg, 1175 μg, 1200 μg, 1225 μg, 1250 μg, 1275 μg, 1300 μg, 1325 μg, 1350 μg, 1375 μg, 1400 μg, 1425 μg, 1450 μg, 1475 μg, 1500 μg, 1525 μg, 1550 μg, 1575 μg, 1600 pg , 1625 μg, 1650 μg, 1675 μg, 1700 μg, 1725 μg, 1750 μg, 1775 μg, 1800 μg, 1825 μg, 1850 μg, 1875 μg, 1900 μg, 1925 μg, 1950 μg, 1975 μg, 2000 μg, 2025 μg, 2050 μg, 2075 μg, 2100 μg, 2125 μg, 2150 μg, 2175 μg, 2200 μg, 2225 μg, 2250 μg, 2275 μg, 2300 μg, 2325 μg, 2350 μg, 2375 μg, 2400 μg, 2425 μg, 2450 μg, 2475 μg, 2500 μg, 2525 μg, 2550 μg, 2575 μg, 2600 μg, 2625 μg, 2650 μg, 2675 μg, 2700 μg, or 2725 μg), and more preferably between about 1000 and 2500 μg (e.g., about 1025 μg, 1050 μg, 1075 μg, 1100 μg, 1125 μg, 1150 μg, 1175 μg, 1200 μg, 1225 μg, 1250 μg, 1275 μg, 1300 μg, 1325 μg, 1350 μg, 1375 μg, 1400 μg, 1425 μg, 1450 μg, 1475 μg, 1500 μg, 1525 μg, 1550 μg, 1575 μg, 1600 μg , 1625 μg, 1650 μg, 1675 μg, 1700 μg, 1725 μg, 1750 μg, 1775 μg, 1800 μg, 1825 μg, 1850 μg, 1875 μg, 1900 μg, 1925 μg, 1950 μg, 1975 μg, 2000 μg, 2025 μg, 2050 μg, 2075 μg, 2100 μg, 2125 μg, 2150 μg, 2175 μg, 2200 μg, 2225 μg, 2250 μg, 2275 μg, 2300 μg, 2325 μg, 2350 μg, 2375 μg, 2400 μg, 2425 μg, 2450 μg, or 2475 μg), most preferably between about 1000 μg to 2000 μg (e.g., about 1025 μg, 1050 μg, 1075 μg, 1100 μg, 1125 μg, 1150 μg, 1175 μg, 1200 μg, 1225 μg, 1250 μg, 1275 μg, 1300 μg, 1325 μg, 1350 μg, 1375 μg, 1400 μg, 1425 μg, 1450 μg, 1475 μg, 1500 μg, 1525 μg, 1550 μg, 1575 μg, 1600 μg , 1625 μg, 1650 μg, 1675 μg, 1700 μg, 1725 μg, 1750 μg, 1775 μg, 1800 μg, 1825 μg, 1850 μg, 1875 μg, 1900 μg, 1925 μg, 1950 μg, or 1975 μg. In some embodiments, the starting dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount between about 1000 μg to 2000 μg. In some embodiments, the starting dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount between about 1500 μg to 2000 μg. In a particular embodiment, the starting dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of at least about 0.25, 0.5, 0.75, 1.0, 1.25 or 1.5 mgs.
[0082] In certain embodiments, administration of the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be repeated prior to the dose titration in step (c) of the methods provided herein. In some embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be repeated for a period of two days, 60 hours, three days, 84 hours, four days, five days, six days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 4 weeks, 6 weeks, 2 months, 75 days, 3 months, or 6 months prior to the dose titration in step (c). In a particular embodiment, the starting dose of the insulinotropic peptide or modified insulinotropic peptide is repeated for 4 weeks prior to the dose titration in step (c).
[0083] In some embodiments, the repeated administrations of the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be separated by at least 12 hours, one day, 36 hours, two days, 60 hours, three days, 84 hours, four days, five days, six days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 4 weeks, 6 weeks, 2 months, 75 days, 3 months, or 6 months. In certain embodiments, the repeated administration of the starting dose of the insulinotropic peptide or modified insulinotropic peptide is separated by three or four days, by one week, or by two weeks.
[0084] In a particular embodiment, administration of the starting dose of the insulinotropic peptide or modified insulinotropic peptide is repeated once every 1, 2, 3, 4, 5, 6 or 7 days for a period of one, two, three, four, or more than four weeks prior to the dose titration of step (c).
[0085] In some embodiments, the repeated administrations of the starting dose of the insulinotropic peptide or modified insulinotropic peptide is administered to the subject in accordance with a weekly dosing regime administered over a number of weeks, before commencement of the dose titration in step (c) of the methods provided herein. In some embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be administered once a week (e.g., as a single dose). In some embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be administered twice a week (e.g., as two of the same or different doses). In other embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be administered once every 2, 3, 4, 5 or 6 days. In other embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be administered once every 8, 9, 10, 11, 12 or 13 days. In other embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be administered two times every 3, 4, 5, 6, 7 or 8 day period. In other embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide can be administered two times every 9, 10, 11, 12, 13 or 14 day period.
[0086] In certain embodiments, the starting dose of the insulinotropic peptide or modified insulinotropic peptide is administered in a single administration during the week, i.e., once a week, and the total weekly dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1000 μg or 1500 μg. In certain embodiments, the total weekly dose is administered once a week, and the dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 2000 μg. In certain embodiments, the total weekly dose is administered once a week, and the dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 2500 μg. In certain embodiments, the total weekly dose is administered once a week, and the dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 3000 μg. In certain embodiments, the total weekly dose is administered once a week, and the dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 3500 μg. In certain embodiments, the total weekly dose is administered once a week, and the dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 4000 μg. In certain embodiments, the total weekly dose is administered once a week, and the dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 4500 μg. In certain embodiments, the total weekly dose is administered once a week, and the dose comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 5000 μg.
[0087] In certain embodiments, the total weekly dose is administered over two administrations during the week, i.e., twice a week, with two equal doses or two unequal doses. In certain embodiments, the total weekly dose is administered twice a week with two unequal doses, wherein the smaller of the two unequal doses is administered prior to the larger of the two unequal doses. In certain embodiments, the total weekly dose is administered twice a week with two unequal doses, wherein the larger of the two unequal doses is administered prior to the smaller of the two unequal doses.
[0088] In certain embodiments, the total weekly dose is administered over two administrations during the week, i.e., twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1000 μg, amounting to a total weekly dose of 2000 μg. In certain embodiments, the total weekly dose is administered twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1500 μg, amounting to a total weekly dose of 3000 μg. In certain embodiments, the total weekly dose is administered twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1600 μg, amounting to a total weekly dose of 3200 μg. In certain embodiments, the total weekly dose is administered twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1700 μg, amounting to a total weekly dose of 3400 μg. In certain embodiments, the total weekly dose is administered twice a week, wherein the first administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1500 μg and the second administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 2000 μg, amounting to a total weekly dose of 3500 μg. In certain embodiments, the total weekly dose is administered twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1750 μg, amounting to a total weekly dose of 3500 μg. In certain embodiments, the total weekly dose is administered twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1800 μg, amounting to a total weekly dose of 3600 μg. In certain embodiments, the total weekly dose is administered twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 1900 μg, amounting to a total weekly dose of 3800 μg. In certain embodiments, the total weekly dose is administered twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 2000 μg, amounting to a total weekly dose of 4000 μg. In certain embodiments, the total weekly dose is administered twice a week, wherein the first administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 2000 μg and the second administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 2500 μg, amounting to a total weekly dose of 4500 μg. In certain embodiments, the total weekly dose is administered twice a week, and each administration comprises the insulinotropic peptide or modified insulinotropic peptide in an amount of 2500 μg, amounting to a total weekly dose of 5000 μ^
C. Dose Titration
[0089] In some embodiments, step (c) of the methods provided herein comprises repeating a cycle comprising the steps of: (i) determining the fasting glucose level of the subject; and (ii) administering to the subject one or more further doses, described herein, of the pharmaceutical formulation comprising the insulinotropic peptide or modified insulinotropic peptide. In some embodiments, the cycle of said determining and administering is repeated until a determining step of a cycle indicates that the fasting blood glucose level of the subject is within the target range. In some embodiments, the cycle of said determining and administering is repeated for at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more cycles. In a particular embodiment, the cycle of said determining and administering is repeated for at least 10 cycles.
[0090] In some embodiments, within each cycle of determining and administering, administration of the dose of the insulinotropic peptide or modified insulinotropic peptide is performed within 24 hours of determining the fasting blood glucose level of the subject. In some embodiments, administration of the dose of the insulinotropic peptide or modified insulinotropic peptide is performed within about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours of determining the fasting blood glucose level of the subject. In a particular embodiment, administration of the dose of the insulinotropic peptide or modified insulinotropic peptide is performed within about 3 hours of determining the fasting blood glucose level of the subject.
[0091] In some embodiments, the dose of the insulinotropic peptide or modified insulinotropic peptide administered over the course of the repeated cycles can be held constant, or can be varied, e.g., increased or decreased, relative to a previously administered dose. In some embodiments, the dose of the insulinotropic peptide or modified insulinotropic peptide administered over the course of the repeated cycles can be incrementally increased or decreased relative to any previously administered dose of the administration regimen. In some embodiments, the dose titration of step (c) may comprise administering a constant dose for a plurality cycles, followed by administering an increased or decreased dose relative to the constant dose for one or more further cycles until a determining step of a cycle indicates that the fasting blood glucose level of the subject is within the target range. In some embodiments, the dose titration of step (c) may comprise administering a constant dose for a plurality cycles, followed by administering an increased or decreased dose relative to any previously administered dose for one or more further cycles until a determining step of a cycle indicates that the fasting blood glucose level of the subject is within the target range. In particular embodiments, administering the dose of the insulinotropic peptide or modified insulinotropic peptide over the course of the repeated cycles comprises incrementally increasing the dose of the insulinotropic peptide or modified insulinotropic peptide relative to a previously administered dose, for example, relative to the immediately preceding dose, until a determining step of a cycle indicates that the fasting blood glucose level of the subject is within the target range.
[0092] In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide that is about 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 μg greater than a previously administered dose, for example, the
immediately preceding dose. In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide that is about 100, 200, 250, 300, 400, 500, 600, 700, 750, 800, 900 or 1000 μg greater than a previously administered dose, for example, the immediately preceding dose. In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide that is over about 1000 μg greater than a previously administered dose, e.g., about 1250, 1500, 1750, 2000, 2250, 2500, 2750, 3000, 3250, 3500, 3750, 4000, 4250, 4500, 4750, 5000 μg, or more, greater than a previously administered dose. In some
embodiments, the administering step of a cycle comprises administering a dose of the
insulinotropic peptide or modified insulinotropic peptide that is greater than a previously administered dose, for example, the immediately preceding dose, in an amount between about 10 μg and 1000 gg (e.g., about 15 μg, 20 μg, 25 μg, 30 μg, 35 μg, 40 μg, 45 μg, 50 μg, 55 μg, 60 μg, 65 μg, 70 μg, 75 μg, 80 μg, 85 μg, 90 μg, 95 μg, 100 μg, 105 μg, 110 μg, 115 μg, 120 μg, 125 μg, 130 μg, 135 μg, 140 μg, 145 μg, 150 μg, 155 μg, 160 μg, 165 μg, 170 μg, 175 μg, 180 μg, 185 μg, 190 μg, 200 μg, 205 μg, 210 μg, 215 μg, 220 μg, 225 μg, 230 μg, 235 μg, 240 μg, 245 μg, 250 μg, 255 μg, 260 μg, 265 μg, 270 μg, 275 μg, 280 μg, 285 μg, 290 μg, 300 μg, 300 μg, 305 μg, 310 μg, 315 μg, 320 μg, 325 μg, 330 μg, 335 μg , 340 μg, 345 μg, 350 μg, 355 μg, 360 μg, 365 μg, 370 μg, 375 μg, 380 μg, 385 μg, 390 μg, 400 μg, 405 μg, 410 μg, 415 μg, 420 μg, 425 μg, 430 μg, 435 μg , 440 μg, 445 μ^ 450 μg, 455 μg, 460 μ^ 465 μg, 470 μ^ 475 μg, 480 μg, 485 μg, 490 μg, 500 μg, 505 μg, 510 μg, 515 μg, 520 μg, 525 μg, 530 μg, 535 μg , 540 μg, 545 μg, 550 μg, 555 μg, 560 μg, 565 μg, 570 μg, 575 μg, 580η, 585 μg, 590 μg, 600 μg, 605 μg, 610 μg, 615 μg, 620 μg, 625 μg, 630 μg, 635 μg , 640 μg, 645 μg, 650 μg, 655 μg, 660 μg, 665 μg, 670 μg, 675 μg, 680 μg, 685 μg, 690 μg, 700 μg, 705 μg, 710 μg, 715 μg, 720 μg, 725 μg, 730 μg, 735 μg , 740 μg, 745 μg, 750 μg, 755 μg, 760 μg, 765 μg, 770 μg, 775 μg, 780 μg, 785 μg, 790 μg, 800 μg, 805 μg, 810 μg, 815 μg, 820 μg, 825 μg, 830 μg, 835 μg , 840 μg, 845 μg, 850 μg, 855 μg, 860 μg, 865 μg, 870 μg, 875 μg, 880 μg, 885 μg, 890 μg, 900 μg, 905 μg, 910 μg, 915 μg, 920 μg, 925 μg, 930 μg, 935 μg, 940 μg, 945 μg, 950 μg, 955 μg, 960 μg, 965 μg, 970 μg, 975 μg, 980 μg, 985 μg, 990 μg or 995 μg of the insulinotropic peptide or modified insulinotropic peptide.
[0093] In other embodiments, administering the dose of the insulinotropic peptide or modified insulinotropic peptide over the course of the repeated cycles comprises incrementally decreasing the dose of the insulinotropic peptide or modified insulinotropic peptide relative to a previously administered dose, for example, relative to the immediately preceding dose, until a determining step of a cycle indicates that the fasting blood glucose level of the subject is within the target range.
[0094] In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide that is 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 μg less than a previously administered dose, for example, the immediately preceding dose. In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide that is about 100, 200, 300, 400, 500, 600, 700, 800, 900 or 1000 μg less than a previously administered dose, for example, the immediately preceding dose. In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide that is over about 1000 μg less than a previously administered dose, e.g., about 1250, 1500, 1750, 2000, 2250, 2500, 2750, 3000, 3250, 3500, 3750, 4000, 4250, 4500, 4750, 5000 μ^ or more, less than a previously administered dose. In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide that is less than a previously administered dose, for example, the immediately preceding dose, in an amount between about 10 μg and 1000 μg (e.g., about 15 μg, 20 μg, 25 μg, 30 μg, 35 μg, 40 μg, 45 μg, 50 μg, 55 μg, 60 μg, 65 μg, 70 μg, 75 μg, 80 μg, 85 μg, 90 μg, 95 μg, 100 μg, 105 μg, 110 μg, 115 μg, 120 μg, 125 μg, 130 μg, 135 μg, 140 μg, 145 μg, 150 μg, 155 μg, 160 μg, 165 μg, 170 μg, 175 μg, 180 μg, 185 μg, 190 μg, 200 μg, 205 μg, 210 μg, 215 μg, 220 μg, 225 μg, 230 μg, 235 μg , 240 μg, 245 μg, 250 μg, 255 μg, 260 μg, 265 μg, 270 μg, 275 μg, 280 μg, 285 μg, 290 μg, 300 μg, 300 μg, 305 μg, 310 μg, 315 μg, 320 μg, 325 μg, 330 μg, 335 μg , 340 μg, 345 μg, 350 μg, 355 μg, 360 μg, 365 μg, 370 μg, 375 μg, 380 μg, 385 μg, 390 μg, 400 μg, 405 μg, 410 μg, 415 μg, 420 μg, 425 μg, 430 μg, 435 μg , 440 μg, 445 μg, 450 μg, 455 μg, 460 μg, 465 μg, 470 μg, 475 μg, 480 μg, 485 μg, 490 μg, 500 μg, 505 μg, 510 μg, 515 μg, 520 μg, 525 μg, 530 μg, 535 μg , 540 μg, 545 μg, 550 μg, 555 μg, 560 μg, 565 μg, 570 μg, 575 μg, 580 μg, 585 μg, 590 μg, 600 μg, 605 μg, 610 μg, 615 μg, 620 μg, 625 μg, 630 μg, 635 μg , 640 μg, 645 μg, 650 μg, 655 μg, 660 μg, 665 μg, 670 μg, 675 μg, 680 μg, 685 μg, 690 μg, 700 μg, 705 μg, 710 μg, 715 μg, 720 μg, 725 μg, 730 μg, 735 μg , 740 μg, 745 μg, 750 μg, 755 μg, 760 μg, 765 μg, 770 μg, 775 μg, 780 μg, 785 μg, 790 μg, 800 μg, 805 μg, 810 μg, 815 μg, 820 μg, 825 μg, 830 μg, 835 μg, 840 μg, 845 μg, 850 μg, 855 μg, 860 μg, 865 μg, 870 μg, 875 μg, 880 μg, 885 μ§, 890 μ§, 900 μ§, 905 μ§, 910 μ§, 915 μ§, 920 μ§, 925 μ§, 930 μ§, 935 μ§ , 940 μ§, 945 μ§, 950 μg, 955 μg, 960 μg, 965 μg, 970 μg, 975 μg, 980 μg, 985 μg, 990 μg or 995 μg of the insulinotropic peptide or modified insulinotropic peptide.
[0095] In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide in an amount of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more milligrams (mgs).
[0096] In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide in an amount between about 500 μg and 3000 μg (e.g., about 525 μg, 550 μg, 575 μg, 600 μg, 625 μg, 650 μg, 675 μg, 700 μg, 725 μg, 750 μg, 775 μg, 800 μg, 825 μg, 850 μg, 875 μg, 900 μg, 925 μg, 950 μg, 975 μg, 1000 μg, 1025 μg, 1050 μg, 1075 μg, 1100 μg, 1125 μg, 1150 μg, 1175 μg, 1200 μg, 1225 μg, 1250 μg, 1275 μg, 1300 μg, 1325 μg, 1350 μg, 1375 μg, 1400 μg, 1425 μg, 1450 μg, 1475 μg, 1500 μg, 1525 μg, 1550 μg, 1575 μg, 1600 μg , 1625 μg, 1650 μg, 1675 μg, 1700 μg, 1725 μg, 1750 μg, 1775 μg, 1800 μg, 1825 μg, 1850 μg, 1875 μg, 1900 μg, 1925 μg, 1950 μg, 1975 μg, 2000 μg, 2025 μg, 2050 μg, 2075 μg, 2100 μg, 2125 μg, 2150 μg, 2175 μg, 2200 μg, 2225 μg, 2250 μg, 2275 μg, 2300 μg, 2325 μg, 2350 μg, 2375 μg, 2400 μg, 2425 μg, 2450 μg, 2475 μg, 2500 μg, 2525 μg, 2550 μg, 2575 μg, 2600 μg, 2625 μg, 2650 μg, 2675 μg, 2700 μg, 2725 μg, 2750 μg, 2775 μg, 2800 μg, 2825 μg, 2850 μ^ 2875 μg, 2900 μ^ 2925 μg, 2950 μg, or 2975 μg).
[0097] In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide in an amount between about 3000 μg and 6000 μg (e.g., about 3025 μg, 3050 μg, 3075 μg, 3100 μg, 3125 μg, 3150 μg, 3175 μg, 3200 μg, 3225 μg, 3250 μg, 3275 μg, 3300 μg, 3325 μg, 3350 μg, 3375 μg, 3400 μg, 3425 μg, 3450 μg, 3475 μg, 3500 μg, 3525 μg, 3550 μg, 3575 μg, 3600 μg , 3625 μg, 3650 μg, 3675 μg, 3700 μg, 3725 μg, 3750 μg, 3775 μg, 3800 μg, 3825 μg, 3850 μg, 3875 μg, 3900 μg, 3925 μg, 3950 μg, 3975 μg, 4000 μ&, 4025 μg, 4050 μg, 4075 μg, 4100 μg, 4125 μg, 4150 μg, 4175 μg, 4200 μg, 4225 μ^ 4250 μg, 4275 μg, 4300 μ^ 4325 μg, 4350 μ^ 4375 μg, 4400 μg, 4425 μ^ 4450 μg, 4475 μg, 4500 μg, 4525 μg, 4550 μg, 4575 μg, 4600 μg , 4625 μg, 4650 μg, 4675 μg, 4700 μg, 4725 μ^ 4750 μg, 4775 μ^ 4800 μg, 4825 μg, 4850 μ^ 4875 μg, 4900 μg, 4925 μg, 4950 μg, 4975 μg, 5000 μg, 5025 μg, 5050 μg, 5075 μg, 5100 μg, 5125 μg, 5250 μg, 5275 μg, 5300 μg, 5325 μg, 5350 μg, 5375 μg, 5400 μg, 5425 μg, 5450 μg, 5475 μg, 5500 μg, 5525 μg, 5550 μg, 5575 μg, 5600 μg , 5625 μg, 5650 μg, 5675 μg, 5700 μg, 5725 μg, 5750 μg, 5775 μg, 5800 μg, 5825 μg, 5850 μg, 5875 μg, 5900 μg, 5925 μg, 5950 μg, or 5975 μg).
[0098] In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide in an amount between about 6000 μg and 9000 μg (e.g., about 6025 μg, 6050 μg, 6075 μg, 6600 μg, 6625 μg, 6650 μg, 6675 μg, 6200 μg, 6225 μg, 6250 μg, 6275 μg, 6300 μg, 6325 μg, 6350 μg, 6375 μg, 6400 μg, 6425 μg, 6450 μg, 6475 μg, 6500 μg, 6525 μg, 6550 μg, 6575 μg, 6600 μg , 6625 μg, 6650 μg, 6675 μg, 6700 μg, 6725 μg, 6750 μg, 6775 μg, 6800 μg, 6825 μg, 6850 μg, 6875 μg, 6900 μg, 6925 μg, 6950 μg, 6975 μg, 7000 μ^ 7025 μg, 7050 μg, 7075 μ^ 7700 μg, 7725 μ^ 7750 μg, 7775 μg, 7200 μg, 7225 μg, 7250 μ^ 7275 μg, 7300 μg, 7325 μ^ 7350 μg, 7375 μ^ 7400 μg, 7425 μg, 7450 μg, 7475 μg, 7500 μg, 7525 μg, 7550 μg, 7575 μg, 7600 μg , 7625 μg, 7650 μg, 7675 μg, 7700 μg, 7725 μg, 7750 μg, 7775 μg, 7800 μg, 7825 μg, 7850 μg, 7875 μg, 7900 μg, 7925 μg, 7950 μg, 7975 μg, 8000 μg, 8025 μg, 8050 μg, 8075 μg, 8800 μg, 8825 μg, 8850 μg, 8875 μg, 8200 μg, 8225 μg, 8250 μg, 8275 μg, 8300 μg, 8325 μg, 8350 μg, 8375 μg, 8400 μg, 8425 μg, 8450 μg, 8475 μg, 8500 μg, 8525 μg, 8550 μg, 8575 μg, 8600 μg , 8625 μg, 8650 μg, 8675 μg, 8700 μg, 8725 μg, 8750 μg, 8775 μg, 8800 μg, 8825 μg, 8850 μg, 8875 μg, 8900 μg, 8925 μg, 8950 μg, or 8975 μg).
[0099] In some embodiments, the administering step of a cycle comprises administering a dose of the insulinotropic peptide or modified insulinotropic peptide in an amount between about 9000 μg and 13,000 μg (e.g., about 9025 μg, 9050 μg, 9075 μ^ 9900 μg, 9925 μg, 9950 μ^ 9975 μ^ 9200 μg, 9225 μg, 9250 μ^ 9275 μg, 9300 μg, 9325 μ^ 9350 μg, 9375 μ^ 9400 μg, 9425 μg, 9450 μg, 9475 μg, 9500 μg, 9525 μg, 9550 μg, 9575 μg, 9600 μg , 9625 9650 μg, 9675 9700 μg, 9725 9750 μ , 9775 μg, 9800 μ^ 9825 μg, 9850 μ^ 9875 μ , 9900 μg, 9925 μ^ 9950 μg, 9975 μ^ 10,000 μ , 10,025 μg, 10,050 μ^ 10,075 μg, 10,10,00 μ , 10,10,25 μ^ 10,10,50 μg, 10,10,75 μ^ 10,200 μg, 10,225 μ , 10,250 μ^ 10,275 μg, 10,300 μ , 10,325 μg,
10,350 μ^ >, 10,375 μg, 10,400 μg , 10,425 μg 10,450 μg 10,475 μg 10,500 μg 10,525 μg,
10,550 μί 10,575 μg, 10,600 μg , 10,625 μ§ ', 10,650 μ§ , 10,675 μ§ ', 10,700 μ§ , 10,725 μg,
10,750 μ^ 1, 10,775 μg, 10,800 μg , 10,825 μg 10,850 μ§ 10,875 μg 10,900 μ§ 10,925 μδ,
10,950 μ^ ¾, 10,975 μ§, 11,000 μg , 11,025 μg 11,050 μ§ 11,075 μg 11,100 μ§ 11,125 μg,
11,150 μ^ 1, 11,175 μg, 11,200 μ§ , 11,225 μg 11,250 μg 11,275 μg 11,300 μg 11,325 μg,
11,350 μί 1, 11,375 μg, 11,400 μ§ , 11,425 μδ 11,450 μg 11,475 μg 11,500 μg 11,525 μg,
11,550 μί 11,575 μg, 11,600 μg , 11,625 μ§ ', 11,650 μ§ , 11,675 μ§ ', 11,700 μ§ , 11,725 μg,
11,750 μ^ 1, 11,775 μg, 11,800 μg , 11,825 μg 11,850 μ§ 11,875 μg 11,900 μ§ 11,925 μ§,
11,950 μ^ ?, 11,975 μ§, 12,000 μg , 12,025 μg 12,050 μg 12,075 μg 12,100 μg 12,125 μg,
12,150 μί 1, 12,175 μg, 12,200 μ§ , 12,225 μg 12,250 μg 12,275 μg 12,300 μg 12,325 μg,
12,350 μί 1, 12,375 μg, 12,400 μ§ , 12,425 μδ 12,450 μg 12,475 μδ 12,500 μg 12,525 μg,
12,550 μί 12,575 μg, 12,600 μg , 12,625 μ§ ', 12,650 μ§ , 12,675 μ§ ', 12,700 μ§ , 12,725 μg,
12,750 μ^ 1, 12,775 μg, 12,800 μg , 12,825 μg 12,850 μg 12,875 μg 12,900 μg 12,925 μ§,
12,950 μί or 12,975 μg).
[0100] [η some embodiments, the dose of the insulinotropic peptide or modified insulinot peptide administered over the course of the repeated cycles does not exceed a predetermined maximum dosage. Within these embodiments, the maximum dosage and maximum frequency of administration of the insulinotropic peptide or modified insulinotropic peptide can be determined by one skilled in the art, and will vary with the nature and severity of the disorder or condition underlying the subject's high blood glucose levels, and the route by which the insulinotropic peptide or modified insulinotropic peptide is administered. The maximum frequency and maximum dosage will also vary according to factors specific for each subject depending on the age, body weight, response, and past medical history of the subject. Thus, in some embodiments, the dose of the insulinotropic peptide or modified insulinotropic peptide administered over the course of the repeated cycles can be incrementally increased relative to any previously administered dose of the administration regimen until a maximum dose is administered, after which continuing administrations of the insulinotropic peptide or modified insulinotropic peptide do not exceed the maximum dose. In particular embodiments, the maximum dose of the dose titration is about 5, 6, 7, 8, 9, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more milligrams (mgs) of the insulinotropic peptide or modified insulinotropic peptide. In particular embodiments, the maximum dose of the dose titration is about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% greater than the maximum tolerated dose (MTD) indicated for the insulinotropic peptide or modified insulinotropic peptide. In particular embodiments, the maximum dose of the dose titration is about 2, 3, 4, 5, 6, 7, 8, 9 or 10 times greater than the maximum tolerated dose (MTD) indicated for the insulinotropic peptide or modified insulinotropic peptide.
[0101] In some embodiments, during repeated cycles of determining the fasting blood glucose levels of the subject and administering to the subject a dose of the pharmaceutical formulation comprising the insulinotropic peptide or modified insulinotropic peptide, two consecutive cycles can be separated by 12 hours, one day, 36 hours, two days, 60 hours, three days, 84 hours, four days, five days, six days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 4 weeks, 1 month, 6 weeks, 2 months, 75 days, 3 months, or 6 months. In certain embodiments, two consecutive cycles of determining and administering are separated by three days, four days, five days, six days, one week, or two weeks. In some embodiments, each cycle of determining and administering is performed once every 1, 2, 3, 4, 5, 6 or 7 days. In other embodiments, each cycle of determining and
administering is performed once every 8, 9, 10, 12, 13 or 14 days. In other embodiments, each cycle of determining and administering is performed once every 21 or 28 days, or once a month. In some embodiments, the cycles of determining and administering are repeated until a determining step of a cycle indicates that the fasting blood glucose level of the subject is within the target range.
D. Maintenance
[0102] In some embodiments, the methods provided herein comprises administering a dose for a plurality of times over a period of time whereby said fasting blood glucose level of the subject is maintained within the target range. Thus, once a determination has been made that the fasting blood glucose levels of the subject falls within the target range, the subject is administered a dose, i.e., a "maintenance dose" of the insulinotropic peptide or modified insulinotropic peptide that maintains the fasting blood glucose levels of the subject within the target range. In some embodiments, a maintenance dose of the insulinotropic peptide or modified insulinotropic peptide is the dose administered to the subject during which it was initially determined that the fasting blood glucose level of the subject fell within the target range. In some embodiments, a maintenance dose is greater than the dose administered to the subject during which it was initially determined that the fasting blood glucose level of the subject fell within the target range. In some embodiments, a maintenance dose is less than the dose administered to the subject during which it was initially determined that the fasting blood glucose level of the subject fell within the target range. In particular embodiments, a maintenance dose is the minimum dose required to maintain the fasting blood glucose level of the subject within the target range.
[0103] In other embodiments, once a fasting blood glucose level of the subject is reduced to within the target range, a maintenance dose may be administered at a reduced frequency relative to the frequency of dosing during the dose titration of step (c) of the methods provided herein. In some embodiments, the maintenance phase of the present methods comprises administering the dose administered to the subject during which it was initially determined that the fasting blood glucose level of the subject fell within the target range; however, as a maintenance dose, this dose is administered at a reduced frequency.
[0104] In some embodiments, a maintenance dose of the insulinotropic peptide or modified insulinotropic peptide is administered once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days over a period of time. In some embodiments, a maintenance dose of the insulinotropic peptide or modified insulinotropic peptide can be administered once a week (e.g., as a single dose). In some embodiments, a maintenance dose of the insulinotropic peptide or modified insulinotropic peptide can be administered twice a week (e.g., as two of the same or different doses). In other embodiments, a maintenance dose of the insulinotropic peptide or modified insulinotropic peptide can be administered once every 2, 3, 4, 5 or 6 days. In other embodiments, a maintenance dose of the insulinotropic peptide or modified insulinotropic peptide can be administered once every 8, 9, 10, 11, 12 or 13 days. In other embodiments, a maintenance dose of the insulinotropic peptide or modified insulinotropic peptide can be administered two times every 3, 4, 5, 6, 7 or 8 day period. In other embodiments, a maintenance dose of the insulinotropic peptide or modified
insulinotropic peptide can be administered two times every 9, 10, 11, 12, 13 or 14 day period.
[0105] In some embodiments, the period of time during which a maintenance dose is administered to the subject will be as long as therapy with the insulinotropic peptide or modified insulinotropic peptide is needed. In some embodiments, the period of time during which a maintenance dose is administered can be in the range of 1 day to an unlimited number of days, i.e., chronic administration. Chronic administration can continue for weeks, months, or years if appropriate according to the judgment of the practitioner of skill in the art. Furthermore, if maintenance doses, in the judgment of the practioner of skill in the art, show tolerability profiles which may not be acceptable, e.g., marked by frequent and severe bouts of nausea and vomiting, the practitioner can reduce the maintenance dose to reduce such profiles.
E. Titration Stops
[0106] In some embodiments, if during step (c), i.e., the dose titration of the administration regimens provided herein, the subject experiences one or more of the following conditions: (i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be within the target range, e.g., below said fasting blood glucose level characteristic of the presence of pre-diabetes or diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject; the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
[0107] With regard to the methods of treating a subject having a blood glucose level characteristic of the presence of diabetes provided herein, the fasting blood glucose level characteristic of the presence of diabetes can be any fasting blood glucose level known in the art to be indicative of the presence of diabetes. For example, in some embodiments, the fasting blood glucose level characteristic of diabetes is provided by the American Diabetes Association, and is in accordance with the Standards of Medical Care in Diabetes as published annually. See, e.g., "Standards of Medical Care in Diabetes - 2010," Diabetes Care 2010 Jan; 33 Suppl. 1 :S11-61, the entirety of which is incorporated by reference herein. In some embodiments, the fasting blood glucose level characteristic of the presence of diabetes is a level of greater than 125 mg/dL. Thus, in some embodiments, the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of diabetes when the fasting blood glucose level of the subject is 125 mg/dL or lower, e.g., about 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 or 125 mg/dL. In some embodiments of the methods provided herein, the fasting blood glucose level of the subject is determined to be below said fasting blood glucose level characteristic of the presence of diabetes when two consecutive determinations of the subject's blood glucose level indicates a fasting blood glucose level of 125 mg/dL or lower, e.g., about 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 or 125 mg/dL. In other embodiments of the methods provided herein, the fasting blood glucose level of the subject is determined to be below said fasting blood glucose level characteristic of the presence of diabetes when the average weekly fasting blood glucose level of the subject is 125 mg/dL or lower, e.g., about 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 or 125 mg/dL. In some embodiments, if the value of the fasting blood glucose level of the subject is determined in an occurrence of a determining step to be within the range of 80 to 125 mg/dL, e.g., e.g., about 80, 85, 90, 95, 100, 105, 110, 115, 120 or 125 mg/dL, the next occurrence of an administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject. [0108] With regard to the methods of treating a subject having a blood glucose level characteristic of the presence of pre-diabetes provided herein, the fasting blood glucose level characteristic of the presence of pre-diabetes can be any fasting blood glucose level known in the art to be indicative of the presence of pre-diabetes. For example, in some embodiments, the fasting blood glucose level characteristic of pre-diabetes is provided by the American Diabetes Association, and is in accordance with the Standards of Medical Care in Diabetes as published annually. See, e.g., "Standards of Medical Care in Diabetes - 2010," Diabetes Care 2010 Jan; 33 Suppl. 1 :S11-61, the entirety of which is incorporated by reference herein. In some embodiments, the fasting blood glucose level characteristic of the presence of pre-diabetes is a level in the range of 100 mg/dL to 125 mg/dL. Thus, in some embodiments, the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of pre-diabetes when the fasting blood glucose level of the subject is below 100 mg/dL, e.g., about 70, 75, 80, 85, 90, 95, 96, 97, 98 or 99 mg/dL. In some embodiments of the methods provided herein, the fasting blood glucose level of the subject is determined to be below said fasting blood glucose level characteristic of the presence of pre- diabetes when two consecutive determinations of the subject's blood glucose level indicates a fasting blood glucose level of below 100 mg/dL, e.g., about 70, 75, 80, 85, 90, 95, 96, 97, 98 or 99 mg/dL. In other embodiments of the methods provided herein, the fasting blood glucose level of the subject is determined to be below said fasting blood glucose level characteristic of the presence of pre-diabetes when the average weekly fasting blood glucose level of the subject is below 100 mg/dL, e.g., about 70, 75, 80, 85, 90, 95, 96, 97, 98 or 99 mg/dL. In some embodiments, if the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 80 to 99 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
[0109] In particular embodiments, if the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 80 mg/dL, e.g., below 79, 78, 77, 76, 75, 74, 73, 72, 71, 70 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject. In particular embodiments, if the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 80 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject. In particular embodiments, if the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 75 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
[0110] In some embodiments, the increased amount can be said to be too large to practically administer to the subject when the formulation volume comprising the insulinotropic peptide or modified insulinotropic peptide to be administered subcutaneously in a single dose exceeds 3 mis. In some embodiments, the increased amount is too large to practically administer when the dose to be administered exceeds 0.5, 0.6, 0.7, 0.75, 0.8, 0.9, 1.0, 1.1, 1.2, 1.25, 1.3, 1.4, 1.5, 1.6, 1.7, 1.75, 1.8, 1.9, 2.0, 2.1, 2.2, 2.25, 2.3, 2.4, 2.5, 2.6, 2.7, 2.75, 2.8, 2.9, or 3 mis. In particular embodiments, the increased amount is too large to practically administer when the dose to be administered exceeds about 1.0 ml. In some embodiments, the dose administered is about any of
0.5, 0.6, 0.7, 0.75, 0.8, 0.9, or 1.0 mis or does not exceed 1.0 ml, the concentration is greater than 10 mg/ml (e.g., 10 mg/ml to about 40 mg/ml), and the dose is administered subcutaneously.
[0111] In some embodiments, if the subject experiences a condition of nausea intolerable to the subject, a condition of vomiting intolerable to the subject, said administering of the step comprises administering the same or a decreased dose relative to a previously administered dose, e.g., the immediately preceding dose, until the subject is no longer experiencing said condition. Upon cessation of the condition, the dose titration of the administration regimen can be resumed accordingly. For example, upon cessation of intolerable nausea and/or intolerable vomiting experienced by the subject during the dose titration, the dose titration may be resumed upward,
1. e., the dose of the insulinotropic peptide or modified insulinotropic peptide may be increased incrementally until a fasting blood glucose level within the target range is reached.
[0112] Nausea as referred to herein can be any unpleasant sensation in the epigastrium, in the back of the throat, or in the abdomen. In certain embodiments, the assessment of whether the nausea is intolerable to the subject is determined by the subjective experience of the subject. In certain embodiments, the assessment of whether the nausea is intolerable to the subject is determined by the subjective experience of the subject combined with objective elements such as pallor, sweating and feeling cold. In some embodiments, nausea intolerable to the subject may culminate in vomiting. In some embodiments, nausea intolerable to the subject may not culminate in vomiting. In some embodiments, nausea intolerable to the subject can be sickness at the stomach, especially when accompanied by a loathing for food and an involuntary impulse to vomit. In some embodiments, nausea intolerable to the subject can be a feeling of sickness or discomfort in the stomach marked by an urge to vomit. In some embodiments, the nausea intolerable to the subject is mild. In some embodiments, the nausea intolerable to the subject is moderate. In some embodiments, the nausea intolerable to the subject is severe. In other embodiments the nausea intolerable to the subject does not interfere with the normal daily life of the subject. In some embodiments the nausea intolerable to the subject interferes with normal daily life. In some embodiments the subject is bedridden because of the nausea.
[0113] In other embodiments, the assessment of whether the nausea is intolerable to the subject is determined according to the judgment of the practitioner in the art according to any technique known to those of skill in the art without limitation. In some embodiments the assessment of nausea is determined according to the Duke Descriptive Scale (DDS). See Laszlo et al, 1981, J. Clin. Phamacol. 2:51S-56S; Cotanch, 1983, Cancer Nurs. 6:277-283; the entirety of which is incorporated by reference herein. The DDS grades nausea on a scale of I to IV, taking into account intensity, severity, and impairment in patient activity for a 24-hour period. In grade I, the subject has experienced no nausea. In grade II, the nausea is mild, with no interference in daily activities. In grade III, the nausea is moderate, with some interference in daily activities. In grade IV, the nausea is severe, and the subject is bedridden with nausea for more than 2 hours.
[0114] In some embodiments the assessment of nausea is determined according to the Visual Analog Scale (VAS). See Bennett et al., 1989, Oncol. Nurs. Forum 16: 175, the entirety of which is incorporated by reference herein. The VAS is a line, usually 100 mm in length, occasionally 150 or 160 mm long, with anchors at each end to indicate the extremes of the nausea experienced by the subject. The low endpoint is to the left in a horizontally oriented scale, and at the base of a vertically oriented scale. Subjects indicate the point on the scale corresponding to the degree of nausea they are experiencing. Investigators score the intensity of the discomfort by measuring the millimeters from the low end of the scale to the mark.
[0115] In some embodiments the assessment of nausea is determined according to the Rhodes Index of Nausea and Vomiting (INV). See Rhodes et al, 1994, Cancer Nurs. 17:45-51 ; Rhodes et al, 1986, Oncol. Nurs. Forum 13:41-47; Rhodes et al, 1988, Appl. Nurs. Res. 1 : 143-144; the entireties of each of which are incorporated by reference herein. The INV measures the individual components of nausea, vomiting, retching, and associated distress. This 8-item, 5-point scale measures a subject's perceived (1) duration of nausea; (2) frequency of nausea; (3) distress from nausea; (4) frequency of vomiting; (5) amount of vomiting; (6) distress from vomiting; and (7) frequency of retching. An exemplary INV item is: "During the last 12 hours, I have not felt any distress from nausea / sickness at my stomach"; "During the last 12 hours I have felt mild distress from nausea or sickness at my stomach"; "During the last 12 hours I have felt moderate distress from nausea or sickness at my stomach"; "During the last 12 hours I have felt great distress from nausea or sickness at my stomach"; "During the last 12 hours I have felt as severe distress from nausea or sickness at my stomach as can be." [0116] In some embodiments the assessment of nausea is determined according to the
Functional Living Index-Emesis (FLIE). See Lindley et al, 1992, Qual. Life Res. 1 :331-340, the entirety of which is incorporated by reference herein. The FLIE is a self-administered
questionnaire which focuses on the effects of nausea and vomiting on physical activities, social and emotional function, and the ability to enjoy food. An exemplary FLIE item is: "How much nausea have you had in the past 3 says?" Each item is answered in a range from 1 to 7, with 9 items for nausea and 9 items for vomiting. A total score is created by adding the responses to the 18 questions. The range of total scores possible is between 18 (all 1 responses on the scale) and 126 (all 7 responses on the scale). Lower scores indicate a more negative impact of nausea and vomiting; higher scores indicate a higher impact.
[0117] In some embodiments the assessment of nausea is determined according to a daily diary of the subject. See Baltzer et al, 1994, Cancer 73:720-723; Buser et al, 1993, Ann. Oncol. 4:475- 479; Sung et al, 1993, J. Clin. Anesthes. 5:22-29; the entireties of each of which are incorporated by reference herein. Diaries have been used for periods ranging from 24 hours to 15 days. This method requires subject self -report and has been correlated to other measures such as observation and the FLIE.
[0118] Although the administration regimens described herein provide for an assessment of tolerability as indicated by nausea or vomiting of the subject, other indicia of tolerability can be utilized within the scope of the methods. For example, tolerability can be determined through evaluation of tolerability parameters known to those of skill in the art, including, but not limited to nausea, vomiting, diarrhea, dyspepsia, abdominal distension and/or pain.
III. Methods of Treatment
[0119] Also provided herein are methods of treating in a subject a disorder or condition treatable with an insulinotropic peptide or modified insulinotropic peptide. In certain
embodiments, the disorder or condition treatable with an insulinotropic peptide or modified insulinotropic peptide is obesity. In certain embodiments, the disorder or condition treatable with an insulinotropic peptide or modified insulinotropic peptide is diabetes. While not wishing to be bound by theory, it is believed that pharmaceutical formulations comprising an insulinotropic peptide or modified insulinotropic peptide administered by the methods provided herein will normalize hyperglycemia through glucose-dependent, insulin-dependent and insulin-independent mechanisms. The pharmaceutical formulations are useful as primary agents for the treatment of type II diabetes mellitus and as adjunctive agents for the treatment of type I diabetes mellitus. In certain embodiments, the disorder or condition treatable with an insulinotropic peptide or modified insulinotropic peptide is type II diabetes. In some embodiments, the insulinotropic peptide or modified insulinotropic peptide is an insulinotropic peptide conjugate. In some embodiments, the insulinotropic peptide conjugate is a conjugate of albumin to exendin-4, or a derivative thereof. In preferred embodiments, the subject is a human.
[0120] The methods provided herein are especially suited for the treatment of subjects with diabetes, both type I and type II, in that the action of the insulinotropic peptide or modified insulinotropic peptide is dependent on the glucose concentration of the blood, and thus the risk of hypoglycemic side effects are greatly reduced over the risks in using current methods of treatment.
[0121] Thus, in certain aspects, provided herein are methods of treating type II diabetes mellitus in a subject, comprising administering to a subject having type II diabetes mellitus a formulation comprising an insulinotropic peptide or modified insulinotropic peptide in accordance with an administration regimen described herein. In some embodiments, the insulinotropic peptide or modified insulinotropic peptide is a conjugate of albumin and an insulinotropic peptide, said insulinotropic peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence. In some embodiments, the formulation comprises: the conjugate at a concentration of about 1 mg/ml to about 100 mg/ml; optionally a buffer; a tonicity modifier; a stabilizer; a surfactant, and optionally a preservative, wherein said formulation has a pH from about 4 to about 8. In certain embodiments, the method comprises administering to a subject having type II diabetes mellitus, in accordance with an administration regimen provided herein, a formulation comprising an exendin-4 derivative, the derivative comprising recombinant human serum albumin cysteine 34 thiol covalently linked to a [2- [242 maleimidopropionamido(ethoxy)ethoxy] acetic acid linker covalently linked to the epsilon amino of the carboxy terminal lysine of exendin-4(l-39)Lys40- NH 2.
[0122] The methods provided herein can also be used for the treatment of subjects with obesity. The methods of the present invention can also be used for the treatment of subjects with any disorder or disease treatable with an insulinotropic peptide.
A. Subjects
[0123] In certain embodiments of the invention, the subject is an animal, for example, a mammal, e.g., a non-human primate. In certain embodiments, the subject is a human. The subject can be a male or female subject. In certain embodiments, the subject is a non-human animal, such as, for instance, a cow, sheep, goat, horse, cat or dog.
[0124] In certain embodiments, the subject is at risk for a disorder or a condition treatable with an insulinotropic peptide or modified insulinotropic peptide including, but not limited to, obesity and type II diabetes. In some embodiments the subject is at risk for obesity. In some
embodiments the subject is at risk for type II diabetes. [0125] In some embodiments, the subject is not healthy. In some embodiments the subject has or suffers from a condition treatable with an insulinotropic peptide or modified insulinotropic peptide including, but not limited to, obesity or type II diabetes.
[0126] In some embodiments, the subject is obese. In some embodiments, the subject is a human and has a Body Mass Index (BMI) of 30 kg/m2 or greater. In some embodiments, the subject is a human and has a BMI between 30 kg/m2 and 35 kg/m2. In some embodiments, the subject is a human and has a BMI of 35 kg/m2 or greater. In some embodiments, the subject is a human and has a BMI of 40 kg/m2 or greater. In some embodiments, the subject weighs more than 120% of the normal weight for its age and height and/or ethnicity.
[0127] In some embodiments, the subject has type II diabetes. In some embodiments, the subject has abnormal glucose levels. In particular embodiments, the subject has a high glucose level. In some embodiments, the subject is a human and has an average whole blood glucose level of 8 mmol/L (138 mg/dl) or greater, and/or an average plasma blood glucose level of 9.0 mmol/L (154 mg/dl) or greater. In some embodiments, the subject is a human and has an average whole blood glucose level between 8 mmol/L (138 mg/dl) and 16 mmol/L (281 mg/dl), and/or an average plasma blood glucose level between 9.0 mmol/L (154 mg/dl) and 17 mmol/L (314 mg/dl). In some embodiments, the subject is a human and has an average whole blood glucose level greater than 16 mmol/L (281 mg/dl), and/or an average plasma blood glucose level greater than 17 mmol/L (314 mg/dl).
[0128] In some embodiments, the subject is a human and has a glycosylated hemoglobin (HbAlc) level of 6.5% or greater. In some embodiments, the subject is a human and has a HbAlc level between 6.5% and 11%. In some embodiments, the subject is human and has a HbAlc level of 11 % or greater.
[0129] In certain embodiments, the subject has a disease, disorder or condition treatable with an insulinotropic peptide or modified insulinotropic peptide. For instance, the subject has Metabolic Syndrome. According to the Third Report of the National Cholesterol Education Program's Adult Treatment Panel (ATPIII), a subject with Metabolic Syndrome has three or more of the following criteria: (1) waist circumference of greater than 102 cm for men and greater than 88 cm for women; (2) serum triglycerides of greater than 1.7 mmol/1; (3) blood pressure of greater than 130/85 mmHg; (4) HDL-cholesterol of less than 1.0 mmol/1 in men and less than 1.3 mmol/1 in women; and (5) serum glucose of greater than 6.1 mmol/1 (greater than 5.6 mmol/1 may be applicable). According to the World Health Organization (WHO), a subject with Metabolic Syndrome has diabetes or impaired fasting glucose (IFG) or impaired glucose tolerance (IGT) or insulin resistance (assessed by clamp studies), plus at least two of the following criteria: (1) waist-to-hip ratio of greater than 0.90 in men or greater than 0.85 in women; (2) serum triglycerides of greater than 1.7 mmol/1 or HDL-cholesterol of less than 0.9 mmol/1 in men and less than 1.0 mmol/1 in women; (3) blood pressure of greater than 140/90 mmHg; (4) urinary albumin excretion rate of greater than 20 micrograms/minute or albumin: creatinine ratio of greater than 30 mg/g. Thus, if a subject meets the criteria defined by either the ATPIII or WHO for Metabolic Syndrome, then the subject has Metabolic Syndrome.
[0130] In some embodiments, the subject has pre-diabetes (e.g., impaired glucose tolerance (IGT) or impaired fasting glucose (IFG)). In some embodiments, the subject has diabetes, e.g., type I diabetes, type II diabetes. In some embodiments, the subject has late autoimmune diabetes in adults ("LADA") also known as late onset autoimmune diabetes of adulthood. In some embodiments, the subject has slow onset type I diabetes. In some embodiments, the subject has type 1.5 diabetes. In some embodiments, the subject has steroid induced diabetes. In some embodiments, the subject has Human Immunodeficiency Virus (HIV) Treatment-Induced Diabetes. In some embodiments, the subject has congenital or HIV-Associated Lipodystrophy ("Fat Redistribution Syndrome") related diabetes. In some embodiments, the subject has a nervous system disorder. In some embodiments, the subject has insulin resistance. In some embodiments, the subject has hypoglycemia unawareness. In some embodiments, the subject has restrictive lung disease. In some embodiments, the subject has a gastrointestinal disorder, e.g., irritable bowel syndrome (IBS), functional dyspepsia, or pain associated with gastrointestinal disorders, e.g., pain associated with IBS and functional dyspepsia. In some embodiments, the subject has inflammatory bowel disease (IBD), e.g., Crohn's disease and ulcerative colitis, or pain associated with IBD. In some embodiments, the subject has hyperglycemia, e.g., hyperglycemia associated with surgery (e.g., a major surgical procedure, e.g., coronary bypass surgery) e.g., hyperglycemia associated with surgery on subjects with diabetes, e.g., type II diabetes or metabolic syndrome. In some embodiments, the subject has coronary heart failure (CHF). In some embodiments, the subject has a disorder associated with beta cell dysfunction, disorders associated with the absence of beta cells, or disorders associated with an insufficient number of beta cells. In some embodiments, the subject has a condition or disorder associated with toxic hypervolemia, such as renal failure, congestive heart failure, nephrotic syndrome, cirrhosis, pulmonary edema, and hypertension. In some embodiments, the subject is in need for altering his or her lipoprotein particle size and subclass composition or his or her concentration in fibrinogen.
[0131] In some embodiments, the subject is obese. In some embodiments, the subject is obese but neither diabetic nor pre-diabetic; obese and diabetic or pre-diabetic; obese but not affected by metabolic syndrome; obese and affected by the metabolic syndrome; overweight but neither diabetic nor pre-diabetic; overweight and diabetic or pre-diabetic; overweight but not affected by metabolic syndrome; overweight and affected by metabolic syndrome; affected by metabolic syndrome but neither diabetic nor pre-diabetic (depending on the definition of metabolic syndrome); affected by metabolic syndrome but neither obese nor overweight.
[0132] In some embodiments, the subject has one or more of the following characteristics: (1) diabetes or pre-diabetes; (2) overweight or obesity; and (3) metabolic syndrome.
[0133] In some embodiments, the subject is naive to anti-diabetic agents. In some
embodiments, the subject is naive to other anti-diabetic agents or naive to oral anti-diabetic agents (OAD). In other embodiments, the subject has been previously treated with one or more other antidiabetic agents, e.g., an OAD. In other embodiments, the subject has been previously treated with metformin, a sulfonylurea, a thiazolidinedione or a combination thereof. In some embodiments, the subject is being treated with, i.e., on an active treatment regimen with an OAD. In one embodiment, the subject has been administered an OAD, e.g., metformin within 1 week, 2 days, or 1 day prior to the administration of the insulinotropic peptide conjugate. In a specific embodiment, the subject has been on a stable dose of > 1000 mg metformin daily for at least 3 months. Exemplary OADs are provided below.
[0134] In a particular embodiment, the subject is currently being treated with, i.e., on an active treatment regimen with metformin. In one embodiment, the subject has been administered metformin within 1 week, 2 days, or 1 day prior to the administration of the insulinotropic peptide conjugate. In a particular embodiment, the subject has been on a stable dose of > 1000 mg metformin daily for at least 3 months.
[0135] In certain embodiments, the formulations herein can be administered as monotherapy. In other words, the formulations of the insulinotropic peptide or modified insulinotropic peptide can be provided as the sole administration of an active agent for treatment of one or more conditions provided herein.
B. Combination Therapies with Antidiabetic Agents
[0136] In the methods and formulations provided herein, an insulinotropic peptide or modified insulinotropic peptide can be used with or combined with one or more second therapeutic agents in the treatment or prevention of diabetes, obesity, or disorders treatable with an insulinotropic peptide or modified insulinotropic peptide. In some embodiments, the combinations of these agents can produce a more effective treatment for such diseases or disorders than with either single treatment alone.
[0137] A formulation provided herein can be combined with a second therapeutic agent by any means deemed suitable by a practitioner of skill in the art. For instance, the formulation can be administered as described herein, and the second therapeutic agent can be administered according to any means and according to any schedule and dose suitable for that agent. Methods of administration, doses, and dose schedules are within the skill of those in the art and can be determined based on knowledge of the second active agent. In certain embodiments, doses and dose schedules can be adjusted for combination therapy by those of skill in the art. The formulation and the second agent need not be administered together. However, in certain embodiments, where suitable, the formulation and the second agent can be administered together where appropriate. In certain embodiments, the formulation can comprise the second agent in addition to the insulinotropic peptide or modified insulinotropic peptide where appropriate.
[0138] One or more second therapeutic ingredients or agents can be used together with an insulinotropic peptide or modified insulinotropic peptide in the methods provided herein. Second therapeutic agents include anti-diabetic agents, including oral-anti-diabetic agents (OADs) or anti-obesity agents.
a. OADs
[0139] Exemplary OADs which find use in the combination therapies provided herein include, but are not limited to, sulfonylureas, e.g. tolbutamide (Orinase), acetohexamide (Dymelor), tolazamide (Tolinase), chlorpropamide, (Diabinese), glipizide (Glucotrol), glyburide (Diabeta, Micronase, Glynase), glibenclamide, glimepiride (Amaryl) or gliclazide (Diamicron); biguanides, e.g. metformin, phenformin or buformin; glinide, e.g., Starlix (nateglinide), Prandin (repaglinide), Glufast (mitiglinide); meglitinides, e.g. repaglinide (Prandin) or nateglinide (Starlix);
thiazolidinediones, e.g. rosiglitazone (Avandia), pioglitazone (Actos) or troglitazone (Rezulin); or Alpha-glucosidase inhibitors, e.g. miglitol (Glyset) or acarbose (Precose/Glucobay).
b. DPP IV Inhibitors
[0140] In some embodiments, the second therapeutic agent which finds use in the combination therapies provided herein is a dipeptidyl peptidase IV inhibitor (DPP IV inhibitor). The DPP-IV inhibitor can be any compound that exhibits inhibition of the enzymatic activity of DPP-IV. Examples of DPP-IV inhibitors are described, for example, in (i) D. J. Drucker, 2003, Exp. Opin. Invest. Drugs, 12:87-100; (ii) K. Augustyns, et al., 2003, Exp. Opin. Ther. Patents, 13:499-510; (iii) C. F. Deacon, et al., 2004, Exp. Opin. Investig. Drugs, 13: 1091-1102; (iv) A. E. Weber, 2004, J. Med. Chem., 47:4135-4141 ; (v) J. J. Hoist, 2004, Exp. Opin. Emerg. Drugs, 9: 155-166; (vi) Augustyns et al., 2005, Expert Opinion On Therapeutic Patents, 15(10): 1387-1407; (vii) Sebokova et al., 2007, Current Topics in Medicinal Chemistry 7:547-555, the contents of each of which are incorporated by reference herein in their entireties.
[0141] Where the DPP IV inhibitor is orally available or orally administered, the DPP IV inhibitor is an OAD as described herein. In other words, OADs can include some or all DPP IV inhibitors described herein. [0142] Specific examples of DPP-IV inhibitors include, but are not limited to, dipeptide derivatives or dipeptide mimetics such as alanine -pyrrolidide, isoleucinethiazolidide, and the pseudosubstrate N-valyl prolyl, O-benzoyl hydroxylamine, as described e.g. in U.S. Pat. Nos. 7,253,172, 7,241,756, 7,238,724, 7,238,720, 7,236,683, 7,235,538, 7,230,074, 7,230,002, 7,229,969, 7,223,573, 7,217,711, 7,208,498, 7,205,409, 7,205,323, 7,196,201, 7,192,952, 7,189,728, 7,186,846, 7, 186,731, 7,183,290, 7,183,280, 7,179,809, 7,169,926, 7,169,806, 7,166,579, 7,157,490, 7,144,886, 7,132,443, 7,125,873, 7,125,863, 7,122,555, 7,115,650, 7,109,192, 7,101,871, 7,098,239, 7,084,120, 7,078,397, 7,078,281, 7,074,794, 7,060,722, 7,053,055, 7,034,039, 7,026,316, 6,911,467, 6,890,898, 6,890,905, 6,869,947, 6,867,205, 6,861,440, 6,844,316, 6,849,622, 6,825,169, 6,812,350, 6,803,357, 6,800,650, 6,727,261, 6,716,843, 6,710,040, 6,706,742, 6,699,871, 6,645,995, 6,617,340, 6,699,871, 6,573,287, 6,432,969, 6,395,767, 6,380,398, 6,319,893, 6,303,661, 6,242,422, 6,201,132, 6,172,081, 6,166,063, 6,124,305, 6,110,949, 6,107,317, 6,100,234, 6,040,145, 6,011,155, 5,939,560, 5,462,928, the contents of each of which are incorporated by reference herein in their entireties.
[0143] Further examples of DPP-IV inhibitors can be found in U.S. Pat. App. Pub. Nos. 20070172525, 20070185061, 2007016750, 20070149451, 20070142383, 20070142436, 20070123579, 20070112059, 20070105890, 20070098781, 20070093492, 20070082932, 20070082908, 20070072810, 20070072804, 20070072803, 20070060547, 20070049619, 20070049596, 20070021477, 20060293297, 20060281796, 20060281727, 20060276487, 20060276410, 20060270722, 20060270701, 20060270679,20060264457, 20060264433, 20060264401, 20060264400, 20060258646, 20060258621, 20060247226, 20060229286, 20060217428, 20060211682, 20060205711, 20060205675, 20060173056, 20060154866, 20060142585, 20060135767, 20060135561, 20060135512, 20060116393, 20060111336, 20060111428, 20060079541, 20060074058, 20060074087, 20060069116, 20060058323, 20060052382, 20060046978, 20060040963, 20060039974, 20060014953, 20060014764, 20060004074, 20050059724, 20050059716, 20050043292, 20050038020, 20050032804, 20050272765, 20050272652, 20050261271, 20050260732, 20050260712, 20050245538, 20050234235, 20050233978, 20050234108, 20050222242, 20050222222, 20050222140, 20050215784, 20050215603, 20050209249, 20050209159, 20050203095, 20050203031, 20050203027, 20050192324, 20050187227, 20050176771, 20050171093, 20050164989, 20050143377, 20050143405, 20050137224, 20050131019, 20050130985, 20050130981, 20050113310, 20050107390, 20050107309, 20050096348, 20050090539, 20050075330, 20050070719, 20050070706, 20050070535, 20050070531, 20050070530, 20050065148, 20050065145, 20050065144, 20050043299, 20050043292, 20050032804, 20050026921, 20050004205, 20050004117, 20050032804, 20040259903, 20040259902, 20040259883, 20040259870, 20040259843, 20040254226, 20050254167, 20040242898, 20040242636, 20040242568, 20040242566, 20040236102, 20040235752, 20040229926, 20040229848, 20040229820, 20040209891, 20040186153, 20040180925, 20040176428, 20040176406, 20040171555, 20040171848, 20040167341, 20040167133, 20040152745, 20040147434, 20040138215, 20040138214, 20040121964, 20040116328, 20040110817, 20040106656, 20040106802, 20040106655, 20040097510, 20040087587, 20040082570, 20040082497, 20040077645, 20040072892, 20040063935, 20040034014, 20030232788, 20030225102, 20030216450, 20030216382, 20030199528, 20030195188, 20030166578, 20030162820, 20030149071, 20030134802, 20030130281, 20030130199, 20030125304, 20030119750, 20030119738, 20030105077, 20030100563, 20030092630, 20030087950, 20030078247, 20030060494, 20020198242, 20020198205, 20020183367, 20020165164, 20020161001, 20020110560, 20020103384, 20030096857, 20020071838, 20020065239, 20020061839, 20020049164, 20020019411, 20020006899, 20010020006, the contents of each of which are incorporated by reference herein in their entireties.
[0144] Yet further examples of DPP-IV inhibitors can be found in Application Publication Nos. WO 07/054577, WO 07/053865, WO 05/116029, WO 05/087235, WO 05/082348, WO
05/082849, WO 05/079795, WO 05/075426, WO 05/072530, WO 05/063750, WO 05/058849, WO 05/049022, WO 05/047297, WO 05/044195, WO 05/042488, WO 05/042003, WO
05/040095, WO 05/037828, WO 05/037779, WO 05/034940, WO 05/033099, WO 05/032590, WO 05/030751, WO 05/030127, WO 05/026148, WO 05/025554, WO 05/023762, WO
05/020920, WO 05/19168, WO 05/12312, WO 05/12308, WO 05/12249, WO 05/11581, WO 05/09956, WO 05/03135, WO 05/00848, WO 05/00846, WO 04/112701, WO 04/111051, WO 04/111041, WO 04/110436, WO 04/110375, WO 04/108730, WO 04/104216, WO 04/104215, WO 04/103993, WO 04/103276, WO 04/99134, WO 04/96806, WO 04/92128, WO 04/87650, WO 04/87053, WO 04/85661, WO 04/85378, WO 04/76434, WO 04/76433, WO 04/71454, WO 04/69162, WO 04/67509, WO 04/64778, WO 04/58266, WO 04/52362, WO 04/52850, WO 04/50022, WO 04/50658, WO 04/48379, WO 04/46106, WO 04/43940, WO 04/41820, WO 04/41795, WO 04/37169, WO 04/37181, WO 04/33455, WO 04/32836, WO 04/20407, WO 04/18469, WO 04/18468, WO 04/18467, WO 04/14860, WO 04/09544, WO 04/07468, WO 04/07446, WO 04/04661, WO 04/00327, WO 03/106456, WO 03/104229, WO 03/101958, WO 03/101448, WO 03/99279, WO 03/95425, WO 03/84940, WO 03/82817, WO 03/80633, WO 03/74500, WO 03/72556, WO 03/72528, WO 03/68757, WO 03/68748, WO 03/57666, WO 03/57144, WO 03/55881, WO 03/45228, WO 03/40174, WO 03/38123, WO 03/37327, WO 03/35067, WO 03/35057, WO 03/24965, WO 03/24942, WO 03/22871, WO 03/15775, WO 03/04498, WO 03/04496, WO 03/02530, WO 03/02596, WO 03/02595, WO 03/02593, WO 03/02553, WO 03/02531, WO 03/00181, WO 03/00180, WO 03/00250, WO 02/83109, WO 02/83128, WO 02/76450, WO 02/68420, WO 02/62764, WO 02/55088, WO 02/51836, WO 02/38541, WO 02/34900, WO 02/30891, WO 02/30890, WO 02/14271, WO 02/02560, WO 01/97808, WO 01/96295, WO 01/81337, WO 01/81304, WO 01/68603, WO 01/55105, WO 01/52825, WO 01/34594, WO 00/71135, WO 00/69868, WO 00/56297, WO 00/56296, WO 00/34241, WO 00/23421, WO 00/10549, WO 99/67278, WO 99/62914, WO 99/61431, WO 99/56753, WO 99/25719, WO 99/16864, WO 98/50066, WO 98/50046, WO 98/19998, WO 98/18763, WO 97/40832, WO 95/29691, WO 95/15309, WO 93/10127, WO 93/08259, WO 91/16339, EP 1517907, EP 1513808, EP 1492777, EP 1490335, EP 1489088, EP 1480961, EP
[0145] 1476435, EP 1476429, EP 1469873, EP 1465891, EP 1463727, EP 1461337, EP 1450794, EP 1446116, EP 1442049, EP 1441719, EP 1426366, EP 1412357, EP1406873, EP 1406872, EP 1406622, EP 1404675, EP 1399420, EP 1399471, EP 1399470, EP 1399469, EP 1399433, EP 1399154, EP 1385508, EP 1377288, EP 1355886, EP 1354882, EP 1338592, EP 1333025, EP 1304327, EP 1301187, EP 1296974, EP 1280797, EP 1282600, EP 1261586, EP 1258476, EP 1254113, EP 1248604, EP 1245568, EP 1215207, EP 1228061, EP 1137635, EP 1123272, EP 1104293, EP 1082314, EP 1050540, EP 1043328, EP 0995440, EP 0980249, EP 0975359, EP 0731789, EP 0641347, EP 0610317, EP 0528858, CA 2466870, CA 2433090, CA 2339537, CA 2289125, CA 2289124, CA 2123128, DD 296075, DE 19834591, DE 19828113, DE 19823831, DE 19616486, DE 10333935, DE 10327439, DE 10256264, DE 10251927, DE 10238477, DE 10238470, DE 10238243, DE 10143840, FR 2824825, FR 2822826,
JP2005507261; JP 2005505531, JP 2005502624, JP 2005500321, JP 2005500308,
JP2005023038, JP 2004536115, JP 2004535445, JP 2004535433, JP 2004534836, JP
2004534815, JP 2004532220, JP 2004530729, JP 2004525929, JP 2004525179, JP 2004522786, JP 2004521149, JP 2004503531, JP 2004315496, JP 2004244412, JP 2004043429, JP
2004035574, JP 2004026820, JP 2004026678, JP 2004002368, JP 2004002367, JP 2003535898, JP 2003535034, JP 2003531204, JP 2003531191, JP 2003531118, JP 2003524591, JP
2003520849, JP 2003327532, JP 2003300977, JP 2003238566, JP 2002531547, JP 2002527504, JP 2002517401, JP 2002516318, JP 2002363157, JP 2002356472, JP 2002356471, JP
2002265439, JP 2001510442, JP 2000511559, JP 2000327689, JP 2000191616, JP 1998182613, JP 1998081666, JP 1997509921, JP 1995501078, JP 1993508624, the contents of each of which are incorporated by reference herein in their entireties.
[0146] In certain embodiments, the DPP-IV inhibitor is a small molecule with a molecular weight of less than 1000, 700 or 500 Daltons, e.g., an organic molecule other than a nucleic acid, or a protein or peptide. [0147] In certain embodiments, the DPP-IV inhibitor is a β-aminoacid derivative, such as 3(R)- Amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a- ]pyrazin-7-yl]-4-(2,4,5- trifluorophenyl)butan-l-one (MK-0431 ; sitagliptin; Januvia), or its pharmaceutical salt, hydrate or polymorph, which are described in detail in U.S. Pat. No. 6,699,871, EP 1412357, WO 03/04498, and US 2003100563, the contents of each of which are incorporated by reference herein in their entireties. In some embodiments, the DPP-IV inhibitor is sitagliptin. Sitagliptin is described as an orally active and selective DPP-IV inhibitor and was recently approved in the U.S. and in Europe for the treatment of diabetes alone or in combination with metformin or sulfonylurea or a PPARy agonist. See U.S. Pat. No. 6,699,871, Kim et al., 2005, J. Med. Chem. 48: 141-151, the contents of each of which are incorporated by reference herein in their entireties.
[0148] In certain embodiments, the DPP-IV inhibitor is cyanopyrrolidide, such as (l-[[3- hydroxy-l-adamantyl)amino]acetyl]-2-cyano-(S)-pyrrolidine (LAF237 or vildagliptin; Galvus), 1- [2-[5-cyanopyridin-2-yl)amino]ethylamino]acetyl-2-cyano-(S)-pyrrolidine (NVPDPP728), or (lS,3S,5S)-2- [2 (S)-Amino-2-(3-hydroxyadamantan-l -yl)acetyl]-2-azabicyclo[-3 .1.0]hexane-3- carbonitrile (saxagliptin or BMS-47718; Onglyza), which are disclosed in detail, for example, in U.S. Patent Nos. 6,617,340, 6,432,969, 6,395,767, 6,166,063, 6,124,305, 6,110,949, 6,011,155, 6,107,317, WO 98/19998 and JP 2000511559, WO 00/34241, EP 1137635, and JP 2002531547, the contents of each of which are incorporated by reference herein in their entireties.
[0149] In some embodiments, the DPP-IV inhibitor is vildagliptin. In some embodiments, the DPP-IV inhibitor is NVP-DPP728. Vildagliptin and NVP-DPP728 are described as an orally active and selective DPP-IV inhibitor. See Villhauer et al, 2002, J Med Chem 45:2362-2365, Villhauer et al, 2003, J Med Chem 46:2774-2789, the contents of each of which are incorporated by reference herein in their entireties. Vildagliptin (LAF 237) is currently undergoing Phase III clinical trial in the United States. It is approved for use in Europe in combination in combination with metformin or sulfonylurea or a thiazolidinedione.
[0150] In certain embodiments, the DPP-IV inhibitor is saxagliptin. Saxagliptin is currently in Phase III clinical trail in the U.S. and Europe for the treatment of type II diabetes. See Augeri et al., 2005, J. Med. Chem. 48(5):5025-5037, the contents of which is incorporated by reference herein in its entirety.
[0151] In certain embodiments, the DPP-IV inhibitor is 3-(L-Isoleucyl)thiazolidine (isoleucine- thiazolidide or PSN-9301). Isoleucine-thiazolidide can be found in JP 2001510442, WO
97/40832, U.S. Pat. No. 6,303,661, and DE 19616486, the contents of each of which are incorporated by reference herein in their entireties. Isoleucine-thiazolidide is described as an orally active and selective DPP-IV inhibitor. See Pederson et al, 1998, Diabetes 47: 1253-1258; Epstein et al., 2007, Curr. Opion. Investig. Drugs, 8(4):331-337, the contents of each of which are incorporated by reference herein in their entireties.
[0152] In certain embodiments, the DPP-IV inhibitor is SYR-322 (TAK-622; alogliptin) or SYR-472 such as described in U.S. Pat. Nos. 7, 169,926 and 7,034,039, the contents of each of which are incorporated by reference herein in their entireties.
[0153] In certain embodiments, the DPP-IV inhibitor is valine -pyrrolidide, such as disclosed in Deacon et al, Diabetes (1998) 47:764769; which is incorporated by reference herein in its entirety.
[0154] In certain embodiments, the DPP-IV inhibitor is [l-[2(S)-Amino-3- methylbutyryl]pyrrolidin-2(R)-yl]boronic acid (PT-100).
[0155] In certain embodiments, the DPP-IV inhibitor is (3-phenethylamine, such as described in Nordhoff et al., 2006, Bioorganic Medical Chemistry Letters 16: 1744-1748, is incorporated by reference herein in its entirety.
[0156] In certain embodiments, the DPP-IV inhibitor is PT-630 (DB-160), such as described in Application Publication No. WO 06/034435, which is incorporated by reference herein in its entirety.
[0157] In certain embodiments, the DPP-IV inhibitor is ABT-341, such as described in Pei et al, J. Med. Chem. 2006 Nov 2; 49(22):6439-42, which is incorporated by reference herein in its entirety.
[0158] In certain embodiments, the DPP-IV inhibitor is ABT-279, such as described in Madar et al, J. Med. Chem. 2006 Oct 19; 49(21):6416-20, which is incorporated by reference herein in its entirety.
[0159] In certain embodiments, the DPP-IV inhibitor is BI-1356 (Ondero, linagliptin) such as described in Application Publication No. WO 04/18468, which is incorporated by reference herein in its entirety.
[0160] In certain embodiments, the DPP-IV inhibitor is SYR-619; TAK-619.
[0161] In certain embodiments, the DPP-IV inhibitor is GSK-823093.
[0162] In certain embodiments, the DPP-IV inhibitor is PSN 9301.
[0163] In certain embodiments, the DPP-IV inhibitor is TA-6666.
[0164] In certain embodiments, the DPP-IV inhibitor is CR- 14023.
[0165] In certain embodiments, the DPP-IV inhibitor is CR-14025.
[0166] In certain embodiments, the DPP-IV inhibitor is CR- 14240.
[0167] In certain embodiments, the DPP-IV inhibitor is CR- 13651.
[0168] In certain embodiments, the DPP-IV inhibitor is NNC-72-2138.
[0169] In certain embodiments, the DPP-IV inhibitor is NN-7201. [0170] In certain embodiments, the DPP -IV inhibitor is PHX-1149; dutogliptin.
[0171] In certain embodiments, the DPP -IV inhibitor is PHX-1004.
[0172] In certain embodiments, the DPP- -IV inhibitor is SNT-189379.
[0173] In certain embodiments, the DPP -IV inhibitor is GRC-8087.
[0174] In certain embodiments, the DPP -IV inhibitor is SK-0403.
[0175] In certain embodiments, the DPP -IV inhibitor is GSK-825964.
[0176] In certain embodiments, the DPP -IV inhibitor is TS-021.
[0177] In certain embodiments, the DPP -IV inhibitor is GRC-8200; melogliptin.
[0178] In certain embodiments, the DPP- -IV inhibitor is GRC-8116.
[0179] In certain embodiments, the DPP- -IV inhibitor is FE 107542.
[0180] In certain embodiments, the DPP- -IV inhibitor is MP-513; teneligliptin.
[0181] In certain embodiments, the DPP- -IV inhibitor is BI356.
[0182] In certain embodiments, the DPP -IV inhibitor is ALS 2-0426; AMG-222.
[0183] In certain embodiments, the DPP -IV inhibitor is ABT279.
[0184] In certain embodiments, the DPP -IV inhibitor is TS-201.
[0185] In certain embodiments, the DPP- -IV inhibitor is KRP-104.
[0186] In certain embodiments, the DPP- -IV inhibitor is R1579; carmegliptin.
[0187] In certain embodiments, the DPP -IV inhibitor is LY2463665.
[0188] In certain embodiments, the DPP -IV inhibitor is ARI-2243.
[0189] In certain embodiments, the DPP- -IV inhibitor is SSR-162369.
c. Other Second Therapeutic Agents
[0190] In some embodiments the second therapeutic agent is an insulin receptor agonist. In some embodiments, the insulin receptor agonist is human insulin or insulin analog; basal insulin such as Lantus (insulin glargine), Levemir (insulin detemir), insulin degludec (SIBA; NN1250), insulin degludec plus (insulin degludec + aspart mix; SIAC; NN5401), NN-344, LY-2,605,541, SuliXen (PolyXen insulin), SAR-161,271, InsuLAR (ProPhase insulin), BIOD-Adjustable Basal insulin, Albulin-G (insulin albumin fusion protein), PC-DAC:insulin, or DACdnsulin; or fast acting insulin such as Novolog (insulin aspart), Humalog (insulin lispro), Apidra (insulin glulisine); or non-injectable formulations of insulin such as Afresa (technosphere inhaled insulin) or Oral-lyn (insulin oral spray).
[0191] In some embodiments, the second therapeutic agent is an amylin receptor agonist such as Symlin (pramlintide).
[0192] In some embodiments, the second therapeutic agent is glucose-dependent insulinotropic peptide/ gastric inhibitory polypeptide (GIP) analog; glucagon receptor (GCGR) antagonist such as BAY-27-9955, Cpd G, or ISIS-325,568; glucocorticoid receptor (GCCR) antagonist such as ISIS-377,131; a chromium and vanadium salt or derivative; llbeta-hydroxysteroid dehydrogenase (llbeta-HSDl and llbeta-HSD2) dehydrogenase and reductase inhibitor such as BVT-3498; a protein tyrosine phosphatase lb (PTP lb) inhibitor; glucose transporter (GLUT) and isoforms (GLUT1, GLUT4) inhibitor; sodium-glucose cotransporter and isoforms (SGLT1, SGLT2) inhibitor such as dapaglifozin, sergilfozin, remogliflozin, BI-10,733, JNJ-28431754/ TA-7284, LX-4211, and AVE-2268; sirtuin (SIRT) and isoforms agonist (SIRT1) such as resveratrol, SRT- 501; a PPAR gamma/ delta agonist; a PPAR alpha/ gamma agonist such as tesaglitasar, muraglitazar, naveglitazar; a fructose-1, 6-bisphosphatase (FBPase) inhibitor, such as CS-917, MB-7803; a glucose-dependent insulinotropic receptor (GDIR, G protein-coupled receptor 119, GPR-119) agonist such as ADP-668; a glucose-dependent insulin secretion by G protein-coupled receptors GPR-40, GPR-120, GPR-109A (HM-74A) agonist; fibroblast growth factor (FGF) and isoforms (FGF-21) analog; presenilins-associated rhomboid-like protein (PSARL) antagonist such as CXS-203; hepatic insulin sensitizing substance (HISS), bone morphogenic protein-9 (BMP-9); osteocalcin; visfatin (nicotinamide phosphoribosyl transferase, Nampt); selective PPAR gamma modulator (SPPARM) such as metaglidasen, MBX-2044; glucokinase (GK) activator such as RO-28-1675; glycogen phosphates (GP) inhibitor such as PSN-357; beta-cell growth factor such as islet neogenesis gene-associated protein (INGAP); CD-3 antagonist such as teplizumab, GAD65 antagonist such as Diamyd, DiaPep277, interleukin-1 inhibitor (IL-1) such as XOMA-052, jun N-terminal kinase (JNK) inhibitor, tolerogen such as NBI-6024, TRX4.
[0193] In some embodiments, the second therapeutic agent is an anti-obesity agent. In some embodiments, the anti-obesity agent is a cannabinoid 1 receptor (CB1R) inverse agonist and antagonist such as Acomplia/ Zimulti (rimonabant), Meridia (Sibutramine), or Xenical (Orlistad).
[0194] In some embodiments, the second therapeutic agent is a gastro-intestinal hormone analog. In some embodiments, the gastro-intestinal hormone analog is a glucagon-like peptide-2 (GLP-2) analog such as Gattex (teduglutide); a peptide YY analog such as PYY(l-36), PYY(3- 36); a pancreatic polypeptide (PP) analog; or a gastrin analog.
d. Routes of Adminstration and Dosage of Combination Therapies
[0195] The insulinotropic peptide or modified insulinotropic peptide and the one or more second therapeutic agents can be administered at essentially the same time, i.e., concurrently, e.g., within the same hour or same day, etc., or at separately staggered times, i.e. sequentially prior to or subsequent to the administration of the other anti-diabetic agent, e.g., on separate days, weeks, etc. The instant methods are therefore to be understood to include all such regimes of
simultaneous or non-simultaneous treatment. In some embodiments, the insulinotropic peptide or modified insulinotropic peptide is administered within 0.1, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 , 11, 12, 13, 14, 15, 16, 17, 18 or more than 18 hours of administration of the other second therapeutic agents. In some embodiments, the insulinotropic peptide or modified insulinotropic peptide is administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 , 11, 12, 13, 14 or more than 14 days of administration of the other second therapeutic agents. In some embodiments, the insulinotropic peptide or modified insulinotropic peptide is administered within 1, 2, 3, 4, 5 or more than 5 weeks of administration of the second therapeutic agents.
[0196] In some embodiments of the combination therapies provided herein, the formulation comprising the insulinotropic peptide or modified insulinotropic peptide will be administered to the subject by subcutaneous injection in accordance with a dosing regime provided herein, e.g., at intervals of between 5, 6, 7, 8 or 9 days or at intervals of between 12, 13, 14, 15 or 16 days. Depending on the disease to be treated and the subject's condition, the particular one or more second therapeutic agents can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, intracerebral ventricular (ICV), intracisternal injection or infusion, subcutaneous injection, or implant), inhalation spray, nasal, vaginal, rectal, sublingual, transdermal or topical routes of administration and can be formulated, alone or together, in suitable dosage unit formulations containing conventional non toxic pharmaceutically acceptable diluents, excipients or carriers appropriate for each route of administration. When the particular second therapeutic agent and the insulinotropic peptide or modified insulinotropic peptide are administered separately, they can be administered by different routes.
[0197] The formulation can be administered at any injection site deemed suitable by the practitioner of skill. In certain embodiments, the formulation is administered in the abdomen, thigh or arm.
[0198] The formulation can be administered at any time deemed suitable by the practitioner of skill. In certain embodiments, the formulation is administered in the morning, before a meal or in the evening prior to sleep, or a combination thereof.
[0199] It will be understood, however, that the specific dose level and frequency of dosage for any particular subject can be varied and will depend upon a variety of factors including the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
[0200] In the event that the subject should experience adverse events in response to one or more agents of the combination therapy provided herein, for example, nausea, vomiting, injection-related skin reaction, hypoglycemia, i.e., blood glucose level , 60 mg/dL (3.3 mmol/L) with clinical signs of hypoglycemia, or any other constitutional symptoms or signs, such as extreme and rapid weight loss, the specific dose level and frequency of dosage for one or more of the agents can be reduced or adjusted according to the judgment of the practitioner of skill in the art. [0201] In a particular embodiment of the combination therapy provided herein, the subject receives the insulinotropic peptide or modified insulinotropic peptide and an OAD, e.g., a biguanide, e.g., metformin. In another particular embodiment, the subject receives the
insulinotropic peptide or modified insulinotropic peptide, and two OADs, e.g., a biguanide, e.g., metformin,, sulfonylurea or a thiazolidinedione, and a second OAD.
C. Selecting Subiects for Treatment
[0202] In one aspect, provided herein are methods of selecting a subject for treatment according to the administration regimens provided herein, comprising identifying a subject that has been previously treated with an anti-diabetic agent. Previous treatments with any antidiabetic agent known in the art can serve as a basis for identifying a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the present methods. Exemplary anti-diabetic agents are provided above. In some embodiments, the anti-diabetic agent is an oral anti-diabetic agent (OAD). In some embodiments, the subject is identified for treatment if the subject has not been previously treated with an antidiabetic agent, e.g., an OAD. In other embodiments, the subject is identified for treatment if the subject has previously been treated with an anti-diabetic agent, e.g., an OAD. Whether a subject has been previously treated with an antidiabetic agent, e.g., an OAD, can be determined according to the judgment of the practitioner in the art. In certain embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide conjugate or formulation provided herein, comprising identifying a subject that has experienced hypoglycemia with the other anti-diabetic agent.
[0203] In certain embodiments, provided herein are methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has undergone previous treatment and experienced weight gain or undesirable weight gain.
[0204] In certain embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has been previously treated with a second active agent, e.g., an OAD such as sulfonylurea, metformin or a thiazolidinedione, the method can further comprise determining whether administration of the anti-diabetic agent resulted in a desirable therapeutic outcome, for example, acceptable control of the subject's glucose levels as determined by a practitioner of skill in the art. Acceptable glycemic control can be indicated by, but limited to, a decrease in whole blood glucose, a decrease in plasma blood glucose, a decrease in interstitial glucose (IG), and/or a decrease in HbAlc levels. In some embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has previously been administered an anti-diabetic agent, e.g., an OAD, e.g., resulting in acceptable control of the subject's glucose levels. In a particular embodiment, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has previously been administered an anti -diabetic agent, e.g., an OAD, not resulting in acceptable control of the subject's glucose levels. The foregoing methods can further comprise administering to the identified subject an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein.
[0205] In some embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has been administered an antidiabetic agent, e.g., an OAD, prior to the first administration of the insulinotropic peptide or modified insulinotropic peptide. In a particular embodiment, the OAD is metformin. In some embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has been administered another antidiabetic agent, e.g., an OAD, not more than 30, 25, 20, 15, 10 or 5 days ago (as measured from the time of the identifying), said method further comprising administering an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein within 30, 25, 20, 15, 10 or 5 days of the administration of the other antidiabetic agent. In a particular embodiment, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has not been administered an effective amount of another antidiabetic agent, e.g., an OAD, and then administering the other antidiabetic agent at the time (e.g. within the same hour or the same day as) of the first administration of the insulinotropic peptide or modified insulinotropic peptide. In other embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has not been administered an effective amount of another antidiabetic agent, e.g., an OAD, and then administering to the subject a first administration of the insulinotropic peptide or modified insulinotropic peptide.
[0206] In another aspect, the present invention provides methods for treating a subject having pre-diabetes, e.g., impaired glucose tolerance (IGT) and/or impaired fasting glucose (IFG), comprising administering to said subject an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, in an amount effective to treat pre-diabetes. In some embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has has IFG and/or IGT. In some embodiments, the methods comprise identifying a subject that has a diagnosis of IFG by a practitioner in the art. In other embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has that has a diagnosis of IGT by a practitioner in the art. In some embodiments, the methods comprise identifying a subject that has 2-hour oral glucose tolerance test levels of > 140 mg/dl (7.8 mmol/1) but < 200 mg/dl (11.1 mmol/1). The foregoing methods can further comprise administering to the identified subject an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein.
[0207] In another aspect, the present invention provides methods for treating a subject who is obese but neither diabetic nor pre-diabetic, comprising administering to said subject an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, in an amount effective to treat obesity. In some embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that is obese but neither diabetic nor pre- diabetic, wherein the methods comprise identifying a subject that has been previously treated with an anti-obesity agent. Previous treatments with any anti-obesity agent known in the art can serve as a basis for selection of a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein. In some embodiments, the anti-obesity agent is Orlistat. In some embodiments, the anti-obesity agent is Sibutramine. In other embodiments, the anti-obesity agent is Liraglutide (NN2211). Liraglutide (NN2211) is a GLP-1 analog having the structure Arg(34)Lys(26)-(N-epsilon-(gamma-Glu(N- alpha-hexadecanoyl))-GLP-l(7-36)-NH2. In some embodiments, the subject is selected for treatment if the subject has not been previously treated with Liraglutide. In other embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has previously been treated with
Liraglutide. The foregoing methods can further comprise administering to the identified subject an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein.
[0208] In certain embodiments, where the subject has been previously treated with Liraglutide, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has previously been administered
Liraglutide resulting in a desirable therapeutic outcome, for example, weight loss amounting to greater than 5% of the subject's baseline weight, as determined by a practitioner of skill. In some embodiments, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has previously been administered Liraglutide resulting in weight loss amounting to greater than 5% of the subject's baseline weight. In a particular embodiment, the present invention provides methods of selecting a subject for treatment with an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, comprising identifying a subject that has previously been administered Liraglutide not resulting in weight loss amounting to greater than 5% of the subject's baseline weight. The foregoing methods can further comprise administering to the identified subject an insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein.
IV. Insulinotropic Peptides
[0209] The invention is directed to methods of administering pharmaceutical formulations comprising an insulinotropic peptide or modified insulinotropic peptide. Useful insulinotropic peptides include, but are not limited to, GLP-1, exendin-3 and exendin-4, and their precursors, derivatives and fragments. Useful insulinotropic peptides also include those disclosed in U.S. Patent Nos. 6,514,500; 6,821,949; 6,887,849; 6,849,714; 6,329,336; 6,924,264; WO 03/103572 and 6,593,295, the contents of each of which are incorporated by reference herein in their entireties.
[0210] In a preferred embodiment, the insulinotropic peptide or modified insulinotropic peptide has a C-terminal amide (CO-NH2).
[0211] In some embodiments, the insulinotropic peptides is GLP-1. In some embodiments, the insulinotropic peptides is a GLP-1 derivative. In some embodiments, the insulinotropic peptides is exendin-3. In some embodiments, the insulinotropic peptides is an exendin-3 derivative. In some embodiments, the insulinotropic peptides is exendin-4. In some embodiments, the insulinotropic peptides is an exendin-4 derivative. In some embodiments, the insulinotropic peptides is exendin-4(l-39)-NH2. In some embodiments, the insulinotropic peptides is exendin- 4(l-39)Lys40-NH2.
[0212] In a preferred embodiment, the modified insulinotropic peptide is exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate.
A. GLP-1 and Its Derivatives
[0213] In certain embodiments, the insulinotropic peptide of the insulinotropic peptide conjugate is GLP-1 or a derivative thereof. The hormone glucagon is known to be synthesized as a high molecular weight precursor molecule which is subsequently proteolytic ally cleaved into three peptides: glucagon, glucagon-like peptide 1 (GLP-1), and glucagon-like peptide 2 (GLP-2). GLP-1 has 37 amino acids in its unprocessed form as shown in SEQ ID NO: 1 (HDEFERHAEG TFTSDVSSYL EGQAAKEFIA WLVKGRG). Unprocessed GLP-1 is essentially unable to mediate the induction of insulin biosynthesis. The unprocessed GLP-1 peptide is, however, naturally converted to a 31 -amino acid long peptide (7-37 peptide) having amino acids 7-37 of GLP-1 ("GLP-1 (7-37)") SEQ ID NO:2 (HAEG TFTSDVSSYL EGQAAKEFIA WLVKGRG). GLP-1 (7-37) can also undergo additional processing by proteolytic removal of the C-terminal glycine to produce GLP-1 (7-36) which also exists predominantly with the C-terminal residue, arginine, in amidated form as arginineamide, GLP-1 (7-36) amide. This processing occurs in the intestine and to a much lesser extent in the pancreas, and results in a polypeptide with the insulinotropic activity of GLP-1 (7-37). Thus, GLP-1 peptides useful in the methods and formulations described herein include, but are not limited to, GLP-l(l-37), GLP-1 (7-37), GLP- 1(7-36), and derivatives thereof.
[0214] A compound is said to have an "insulinotropic activity" if it is able to stimulate, or cause the stimulation of, the synthesis or expression of the hormone insulin. The hormonal activity of GLP-1 (7-37) and GLP-1 (7-36) appear to be specific for the pancreatic beta cells where it appears to induce the biosynthesis of insulin. Glucagon-like-peptide hormones are useful in the study of the pathogenesis of maturity onset diabetes mellitus, a condition characterized by hyperglycemia in which the dynamics of insulin secretion are abnormal. Moreover, glucagon-like peptides are useful in the therapy and treatment of this disease, and in the therapy and treatment of hyperglycemia.
[0215] Peptide moieties (fragments) can be chosen from the determined amino acid sequence of human GLP-1. The interchangeable terms "peptide fragment" and "peptide moiety" are meant to include both synthetic and naturally occurring amino acid sequences derivable from a naturally occurring amino acid sequence.
[0216] The amino acid sequence for GLP-1 has been reported by several researchers. See Lopez, L. C. et al, 1983, Proc. Natl. Acad. Set, USA 80:5485-5489; Bell, G. I. et al, 1983, Nature 302:716-718; Heinrich, G. et al, 1984, Endocrinol. 115:2176-2181. The structure of the preproglucagon mRNA and its corresponding amino acid sequence is well known. The proteolytic processing of the precursor gene product, proglucagon, into glucagon and the two insulinotropic peptides has been characterized. As used herein, the notation of GLP-l(l-37) refers to a GLP-1 polypeptide having all amino acids from 1 (N-terminus) through 37 (C-terminus). Similarly, GLP- 1(7-37) refers to a GLP-1 polypeptide having all amino acids from 7 (N- terminus) through 37 (C-terminus). Similarly, GLP- 1(7-36) refers to a GLP-1 polypeptide having all amino acids from number 7 (N-terminus) through number 36 (C-terminus).
[0217] In one embodiment, GLP- 1(7-36) and its peptide fragments are synthesized by conventional means as detailed below, such as by the well-known solid-phase peptide synthesis described by Merrifield, J. M., 1962, Chem. Soc. 85:2149, and Stewart and Young, Solid Phase Peptide Synthesis, Freeman, San Francisco, 1969, pp. 27-66), the contents of each of which are incorporated by reference herein in their entireties. However, it is also possible to obtain fragments of the proglucagon polypeptide, or of GLP-1, by fragmenting the naturally occurring amino acid sequence, using, for example, a proteolytic enzyme. Further, it is possible to obtain the desired fragments of the proglucagon peptide or of GLP-1 through the use of recombinant DNA technology, as disclosed by Maniatis, T., et al., Molecular Biology: A Laboratory Manual, Cold Spring Harbor, N.Y., 1982, which is hereby incorporated by reference herein in its entirety.
[0218] Useful GLP-1 derivatives for use in the methods and formulations described herein include peptides which are derivable from GLP-1 such as GLP-l(l-37) and GLP- 1(7-36). A peptide is said to be "derivable from a naturally occurring amino acid sequence" if it can be obtained by fragmenting a naturally occurring sequence, or if it can be synthesized based upon a knowledge of the sequence of the naturally occurring amino acid sequence or of the genetic material (DNA or RNA) which encodes this sequence. In a specific embodiment, a useful derivative which finds use in the methods and formulations described herein comprises a GLP-1 sequence. In one embodiment, a "GLP-1 derivative" has the following characteristics: (1) it shares substantial homology with GLP-1 or a similarly sized fragment of GLP-1; (2) it is capable of functioning as an insulinotropic hormone; and (3) using at least one of the assays provided herein, the derivative has an insulinotropic activity of at least 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of the insulinotropic activity of GLP-1.
[0219] A derivative of GLP-1 is said to share "substantial homology" with GLP-1 if the amino acid sequence of the derivative shares at least 80%, and more preferably at least 90%, and most preferably at least 95% identity to GLP-l(l-37). Percent identity in this context means the percentage of amino acid residues in the candidate sequence that are identical (i.e., the amino acid residues at a given position in the alignment are the same residue) or similar (i.e., the amino acid substitution at a given position in the alignment is a conservative substitution, as discussed above), to the corresponding amino acid residue in the peptide after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence homology. In certain embodiments, a GLP-1 derivative is characterized by its percent sequence identity or percent sequence similarity with the naturally occurring GLP-1 sequence. Sequence homology, including percentages of sequence identity and similarity, are determined using sequence alignment techniques well-known in the art, preferably computer algorithms designed for this purpose, using the default parameters of said computer algorithms or the software packages containing them.
[0220] Useful derivatives also include GLP-1 fragments which, in addition to containing a sequence that is substantially homologous to that of a naturally occurring GLP-1 peptide may contain one or more additional amino acids at their amino and/or their carboxy termini, or internally within said sequence. Thus, useful derivatives include polypeptide fragments of GLP-1 that may contain one or more amino acids that may not be present in a naturally occurring GLP-1 sequence provided that such polypeptides have an insulinotropic activity of at least 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of the insulinotropic activity of GLP-1. The additional amino acids may be D-amino acids or L-amino acids or combinations thereof.
[0221] Useful GLP-1 fragments also include those which, although containing a sequence that is substantially homologous to that of a naturally occurring GLP-1 peptide, lack one or more additional amino acids at their amino and/or their carboxy termini that are naturally found on a GLP-1 peptide. Thus, useful polypeptide fragments of GLP-1 may lack one or more amino acids that are normally present in a naturally occurring GLP-1 sequence provided that such
polypeptides have an insulinotropic activity of at least 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of the insulinotropic activity of GLP-1.
[0222] Useful derivatives further include GLP-1 fragments which are otherwise identical in sequence to that of a naturally occurring GLP-1 peptide but for the addition, deletion or substitution of no more than 5, 4, 3, 2 or 1 amino acids. In certain embodiments, the derivative contains no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 amino addition, deletion, or substitution relative to a naturally occurring GLP-1 sequence. Thus, useful derivatives include polypeptide fragments of a naturally occurring GLP-1 sequence that are identical but for no more than 5, 4, 3, 2, or 1 amino acid additions, deletions or substitutions relative to the naturally occurring GLP-1 sequence, provided that such polypeptides have an insulinotropic activity of at least 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of the insulinotropic activity of the naturally occurring GLP-1 sequence. In certain embodiments, the polypeptide fragments lack one amino acid normally present in a naturally occurring GLP-1 sequence. In some embodiments, the polypeptide fragments lack two amino acids normally present in a naturally occurring GLP-1 sequence. In some embodiments, the polypeptide fragments lack three amino acids normally present in a naturally occurring GLP-1 sequence. In some embodiments, the polypeptide fragments lack four amino acids normally present in a naturally occurring GLP-1 sequence.
[0223] Useful derivatives also include conservative variants of the above-described fragments which have conservative amino acid substitutions (and thus have amino acid sequences which differ from that of the natural sequence) provided that such variants still have an insulinotropic activity. Examples of conservative substitutions include the substitution of one basic residue for another (i.e. Arg for Lys), the substitution of one hydrophobic residue for another (i.e. Leu for He), or the substitution of one aromatic residue for another (i.e. Phe for Tyr), etc. The following six groups each contain amino acids that are conservative substitutions for one another:
Alanine (A), Serine (S), and Threonine (T)
Aspartic acid (D) and Glutamic acid (E)
Asparagine (N) and Glutamine (Q)
Arginine (R) and Lysine (K)
Isoleucine (I), Leucine (L), Methionine (M), and Valine (V)
Phenylalanine (F), Tyrosine (Y), and Tryptophan (W).
[0224] In addition to those GLP-1 derivatives with insulinotropic activity, GLP-1 derivatives which stimulate glucose uptake by cells but do not stimulate insulin expression or secretion are useful for the methods described herein. Such GLP-1 derivatives are described in U.S. Pat. No. 5,574,008, which is hereby incorporated by reference herein in its entirety.
[0225] GLP-1 derivatives which stimulate glucose uptake by cells but do not stimulate insulin expression or secretion which find use in the methods described herein include:
H2N-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Xaa-Gly- Arg-R2 (SEQ ID NO:3);
H2N-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Xaa-Gly- Arg-R2 (SEQ ID NO:4);
H2N-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Xaa- Gly-Arg-R2 (SEQ ID NO:5);
H2N-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val- Xaa-Gly-Arg-R2 (SEQ ID NO:6);
H2N-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu- Val-Xaa-Gly-Arg-R2 (SEQ ID NO:7); H2N-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp- Leu-Val-Xaa-Gly-Arg-R2 (SEQ ID NO: 8);
H2N-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala- Trp-Leu-Val-Xaa-Gly-Arg-R2 (SEQ ID NO:9);
H2N-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile- Ala-Trp-Leu-Val-Xaa-Gly-Arg-R2 (SEQ ID NO: 10);
H2N-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu- Phe-Ile-Ala-Trp-Leu-Val-Xaa-Gly-Arg-R2 (SEQ ID NO: 11);
H2N-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys- Glu-Phe-Ile-Ala-Trp-Leu-Val-Xaa-Gly-Arg-R2 (SEQ ID NO: 12);
H2N-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala- Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Xaa-Gly-Arg-R2 (SEQ ID NO: 13);
H2N-His-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala- Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Xaa-Gly-Arg-R2 (SEQ ID NO: 14); and
H2N-His-D-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala- Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Xaa-Gly-Arg-R2 (SEQ ID NO: 15).
These peptides are C-terminal GLP-1 fragments which do not have insulinotropic activity but which are nonetheless useful for treating diabetes and hyperglycemic conditions as described in U.S. Pat. No. 5,574,008, which is hereby incorporated by reference herein in its entirety.
[0226] An additional GLP-1 derivative which finds use in the methods and formulations described herein includes a GLP-1 /exendin-4 hybrid peptide comprising GLP-1 (7-36) fused to the nine C-terminal amino acids of exendin-4, having the sequence: HAEG TFTSDVSSYL EGQAAKEFIA WLVKGRPSSGAPPPS (SEQ ID NO:28).
[0227] Also useful in the methods and formulations described herein is the GLP-1 analog Victoza (liraglutide, NN2211). Liraglutide is a GLP-1 analog having the structure
Arg(34)Lys(26)-(N-epsilon-(gamma-Glu(N-alpha-hexadecanoyl))-GLP-l(7-36)-NH2, as described in U.S. Patent No. 6,268,343, which is hereby incorporated by reference in its entirety.
[0228] Also useful in the methods and formulations described herein is the GLP-1 analog semaglutide (NN9535). Semaglutide is a GLP-1 analog having the structure N-epsilon 26- { 18- [N-(17-carboxyheptadecanoyl)-L-Y-glutamyl]-10-oxo-3,6,12,15-tetraoxa-9,18- diazaoctadecanoyl}-[8-(2-amino-2-propanoic acid),34-L-arginine] GLP-1 (7-37)-OH.
[0229] Also useful in the methods and formulations described herein is the GLP-1 analog ROSEOIO (GTP-010, LY307161). ROSEOIO is a GLP-1 analog having the structure Val8-GLP- l(7-37)-OH. [0230] Also useful in the methods and formulations described herein is the GLP-1 analog LY2189265. LY2189265 is a long-acting GLP-1 analog consisting of a dipeptidyl peptidase-IV (DPP-IV)-protected GLP-1 analog covalently linked to a human IgG4-Fc heavy chain by a small peptide linker.
[0231] Also useful in the methods and formulations described herein is the GLP-1 analog LY2428757. LY2428757 is a long-acting GLP-1 analog consisting of a GLP-1 analog covalently linked to a polyethylene glycol (PEG) by a small peptide linker.
[0232] Also useful in the methods and formulations described herein is the GLP-1 analog CNTO-3649. CNTO-3649 is a long-acting GLP-1 analog consisting of a GLP-1 analog covalently linked to a IgG-Fc fragment by a small linker.
[0233] Also useful in the methods and formulations described herein is the GLP-1 analog MODI- 1001. MODI- 1001 is a long-acting GLP-1 analog consisting of a GLP-1 analog fused to the carboxy terminal peptide (CTP) of human chorionic gonadotropin (hCG).
[0234] Also useful in the methods and formulations described herein is the GLP-1 analog taspoglutide (R1583/BIM51077). Taspoglutide is a human GLP-1 analogue comprising two amino-acid substitutions in positions 8 and 35 with aminoisobutyric acid.
[0235] Also useful in the methods and formulations described herein is the GLP-1 derivative comprising a fusion protein molecule as follows: [Gly8]GLP-l(7-36)-[Gly8]GLP-l(7-36)-human serum albumin (Syncria, albiglutide), as described in U.S. Patent No. 7,141,547, which is hereby incorporated by reference in its entirety. In some embodiments, the formulations described herein comprise the [Gly8]GLP-l(7-36)-[Gly8]GLP-l(7-36)-human serum albumin molecule without further modification to the molecule, e.g., without the addition of a reactive group and/or the coupling of albumin thereto, as described in Section V, B.
[0236] Additional GLP-1 derivatives which find use in the methods and formulations described herein include the following GLP-1 fusion protein molecules: GLP-1 (7-36)-human serum albumin; human serum albumin-GLP-l(7-36); [Gly8]GLP-l(7-36)-human serum albumin;
human serum albumin-[Gly8]GLP-l(7-36); GLP-1 (7-36)-GLP-l(7-36)-human serum albumin; GLP-1 (9-36)-human serum albumin; and [Gly8]GLP-l(7-36)-GLP-l(7-36)-human serum albumin, as described in U.S. Patent No. 7,141,547, which is hereby incorporated by reference herein in its entirety. In some embodiments, the formulations described herein comprise the GLP- l(7-36)-human serum albumin; human serum albumin-GLP-l(7-36); [Gly8]GLP-l(7-36)-human serum albumin; human serum albumin-[Gly8]GLP-l(7-36); GLP-1 (7-36)-GLP-l(7-36)-human serum albumin; GLP-1 (9-36)-human serum albumin; or the [Gly8]GLP-l(7-36)-GLP-l(7-36)- human serum albumin molecule without further modification to the molecule, e.g., without the addition of a reactive group and/or the coupling of albumin thereto, as described in Section V, B. [0237] An additional GLP-1 derivative which finds use in the methods and formulations described herein includes a GLP-1 /exendin-4/human serum albumin hybrid polypeptide, comprising [Gly8][Glu22]GLP-l(7-36) fused to the eight C-terminal amino acids of exendin-4(l- 39), fused to a linker sequence, fused to human serum albumin, having the sequence:
HGEGTFTSDV SSYLEEQAAK EFIAWLVKGR GSSGAPPPSG GGGGSGGGGS GGGGSDAHKS EVAHRFKDLG EENFKALVLI AFAQYLQQCP FEDHVKLVNE VTEFAKTCVA DESAENCDKS LHTLFGDKLC TVATLRETYG EMADCCAKQE PERNECFLQH KDDNPNLPRL VRPEVDVMCT AFHDNEETFL KKYLYEIARR HPYFYAPELL FFAKRYKAAF TECCQAADKA ACLLPKLDEL RDEGKASSAK QRLKCASLQK FGERAFKAWA VARLSQRFPK AEFAEVSKLV TDLTKVHTEC CHGDLLECAD DRADLAKYIC ENQDSISSKL KECCEKPLLE KSHCIAEVEN DEMPADLPSL AADFVESKDV CKNYAEAKDV FLGMFLYEYA RRHPDYSVVL LLRLAKTYET TLEKCCAAAD PHECYAKVFD EFKPLVEEPQ NLIKQNCELF EQLGEYKFQN ALLVRYTKKV PQVSTPTLVE VSRNLGKVGS KCCKHPEAKR MPCAEDYLSV VLNQLCVLHE KTPVSDRVTK CCTESLVNRR PCFSALEVDE TYVPKEFNAE TFTFHADICT LSEKERQIKK QTALVELVKH KPKATKEQLK AVMDDFAAFV EKCCKADDKE TCFAEEGKKL VAASQAALGL (SEQ ID NO:29), as described in U.S. Patent No. 7,271,149, which is hereby incorporated by reference in its entirety. In some embodiments, the formulations described herein comprise the GLP-1 /exendin-4/human serum albumin hybrid polypeptide without further modification to the polypeptide, e.g., without the addition of a reactive group and/or the coupling of albumin thereto, as described in Section V. B. Exendin-3 and Exendin-4 Peptides and Derivatives
[0238] Exendin-3 and exendin-4 are 39 amino acid peptides (differing at residues 2 and 3) which are approximately 53% homologous to GLP-1 and find use as insulinotropic agents.
Accordingly, peptides useful in the methods and formulations described herein include, but are not limited to, exendin-3, exendin-4, and derivatives thereof.
[0239] The amino acid sequence of exendin-3 is
HSDGTFTSDLSKQMEEEAVRLFIEWLKNGG PSSGAPPPS (SEQ ID NO: 16), and the amino acid sequence of exendin-4 is HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPS (SEQ ID NO: 17).
[0240] Also useful for the formulations described herein are insulinotropic fragments of exendin-4 comprising the amino acid sequences: exendin-4(l-31) desGlu17 Tyr32 (SEQ ID NO: 18) HGEGTFTSDLSKQMEEAVRLFIEWLKNGGPY and exendin-4(l-30) Tyr31 (SEQ ID NO: 19) HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGY. [0241] Also useful is the inhibitory fragment of native exendin-4 comprising the amino acid sequence: exendin-4(9-39) (SEQ ID NO:20) DLS KQMEEE A VRLFIE WLKNGGPS S GAPPPS .
[0242] Other exemplary insulinotropic peptides useful in the methods and formulations provided herein are presented in SEQ ID NOS:21-27.
HDEFERHAEGTFTSDVSSYLEGQAAKEFIAWLVKGRK SEQ ID NO: 21
HAEGTFTSDVSSYLEGQAAKEFIAWLVKGRK SEQ ID NO: 22
HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPSK SEQ ID NO: 23
HSDGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPSK SEQ ID NO: 24
HGEGTFTSDLSKEMEEEVRLFIEWLKNGGPY SEQ ID NO: 25
HGEGTFTSDLSKEMEEEVRLFIEWLKNGGY SEQ ID NO: 26
DLSKQMEEE A VRLFIE WLKGGPS S GPPPS SEQ ID NO: 27
[0243] Useful exendin-3 and exendin-4 derivatives for use in the methods and formulations described herein also include peptides which are derivable from the naturally occurring exendin-3 and exendin-4 peptides. A peptide is said to be "derivable from a naturally occurring amino acid sequence" if it can be obtained by fragmenting a naturally occurring sequence, or if it can be synthesized based upon a knowledge of the sequence of the naturally occurring amino acid sequence or of the genetic material (DNA or RNA) which encodes this sequence. In a specific embodiment, a useful derivative comprises an exendin-3 or exendin-4 sequence. In one embodiment, an "exendin-3 or exendin-4 derivative" has the following characteristics: (1) it shares substantial homology with exendin-3 or exendin-4 or a similarly sized fragment of exendin-3 or exendin-4; (2) it is capable of functioning as an insulinotropic hormone and (3) using at least one of the assays provided herein, the derivative has an insulinotropic activity of at least 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of the insulinotropic activity of either exendin-3 or exendin-4.
[0244] A derivative of exendin-3 or exendin-4 is said to share "substantial homology" with exendin-3 and exendin-4 if the amino acid sequences of the derivative shares at least 80%, and more preferably at least 90%, and most preferably at least 95% identity to exendin-3 and exendin- 4. Percent identity in this context means the percentage of amino acid residues in the candidate sequence that are identical (i.e., the amino acid residues at a given position in the alignment are the same residue) or similar (i.e., the amino acid substitution at a given position in the alignment is a conservative substitution, as discussed above), to the corresponding amino acid residue in the native peptide after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence homology. In certain embodiments, a exendin-3 or exendin-4 derivative is characterized by its percent sequence identity or percent sequence similarity with the naturally occurring exendin-3 or exendin-4 sequence. Sequence homology, including percentages of sequence identity and similarity, are determined using sequence alignment techniques well- known in the art, preferably computer algorithms designed for this purpose, e.g., BLAST, using the default parameters of said computer algorithms or the software packages containing them.
[0245] Useful derivatives also include exendin-3 or exendin-4 fragments which, in addition to containing a sequence that is the same or that is substantially homologous to that of a naturally occurring exendin-3 or exendin-4 peptide may contain one or more additional amino acids at their amino and/or their carboxy termini, or internally within said sequence. Thus, useful derivatives include polypeptide fragments of exendin-3 or exendin-4 that may contain one or more amino acids that may not be present in a naturally occurring exendin-3 or exendin-4 sequences provided that such polypeptides have an insulinotropic activity of at least 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of the insulinotropic activity of either exendin-3 or exendin-4.
[0246] Similarly, useful derivatives include exendin-3 or exendin-4 fragments which, although containing a sequence that is substantially homologous to that of a naturally occurring exendin-3 or exendin-4 peptide may lack one or more additional amino acids at their amino and/or their carboxy termini that are naturally found on a exendin-3 or exendin-4 peptide. Thus, useful derivatives include polypeptide fragments of exendin-3 or exendin-4 that may lack one or more amino acids that are normally present in a naturally occurring exendin-3 or exendin-4 sequence provided that such polypeptides have an insulinotropic activity of at least 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of the insulinotropic activity of either exendin-3 or exendin-4.
[0247] Useful derivatives further include exendin-3 or exendin-4 fragments which are otherwise identical in sequence to that of the naturally occurring exendin-3 or exendin-4 peptide but for the addition, deletion or substitution of no more than 5, 4, 3, 2 or 1 amino acids. In certain embodiments, the derivative contains no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 amino addition, deletion, or substitution relative to the native exendin-3 or exendin-4 sequence. Thus, useful derivatives include polypeptide fragments of exendin-3 or exendin-4 that are identical but for no more than 5, 4, 3, 2, or 1 amino acid additions, deletions or substitutions relative to the native exendin-3 or exendin-4 sequence, provided that such polypeptides have an insulinotropic activity of at least 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of the insulinotropic activity of either exendin-3 or exendin-4.
[0248] Useful derivatives also include conservative variants of the above-described fragments which have inconsequential amino acid substitutions (and thus have amino acid sequences which differ from that of the natural sequence) provided that such variants still have an insulinotropic activity. Examples of conservative substitutions include the substitution of one basic residue for another (i.e. Arg for Lys), the substitution of one hydrophobic residue for another (i.e. Leu for He), or the substitution of one aromatic residue for another (i.e. Phe for Tyr), etc. The following six groups each contain amino acids that are conservative substitutions for one another:
Alanine (A), Serine (S), and Threonine (T)
Aspartic acid (D) and Glutamic acid (E)
Asparagine (N) and Glutamine (Q)
Arginine (R) and Lysine (K)
Isoleucine (I), Leucine (L), Methionine (M), and Valine (V) Phenylalanine (F), Tyrosine (Y), and Tryptophan (W).
[0249] Also useful in the methods and formulations described herein are the exendin-4 derivatives comprising a fusion protein molecule as follows: exendin-4(l-39)-human serum albumin, and human serum albumin-exendin-4(l-39), as described in U.S. Patent No. 7,141,547 or 7,271,149, the contents of each of which are incorporated by reference herein in their entireties. In some embodiments, the formulations described herein comprise the exendin-4(l- 39)-human serum albumin or human serum albumin-exendin-4(l-39) molecule without further modification to the molecule, e.g., without the addition of a reactive group and/or the coupling of albumin thereto, as described in Section V.
[0250] Also useful in the methods and formulations described herein is the exendin-4 analog lixesenatide (AVE-OOlO/ ZP-10). Lixisenatide is an exendin-4 analog having the structure desPro38-exendin-4[l-39]-Lys x6-NH2 described in U.S. Patent No. 6,528,486, which is hereby incorporated by reference in its entirety.
[0251] Also useful in the methods and formulations described herein is the exendin-4 analog
PF-4856883/ CVX-096. PF-4856883/ CVX-096 is a long-acting exendin-4 analog consisting of an exendin-4 analog covalently linked to an antibody scaffold by a small linker.
[0252] Also useful in the methods and formulations described herein is the exendin-4 analog
LAPS-exendin (HM11260). HM11260 is a long-acting exendin-4 analog consisting of an exendin-4 analog covalently linked to an antibody scaffold by a small linker.
[0253] Also useful in the methods and formulations described herein is the exendin-4 analog exenatide-XTEN (VRS859). VRS859 is a long-acting exendin-4 analog consisting of an exendin-
4 analog covalently linked to a pseudo-PEG protein.
V. Conjugates to Albumin
[0254] Modified insulinotropic peptides useful for the methods described herein include insulinotropic peptides and their derivatives conjugated to albumin. Several methods can be used to link an insulinotropic peptide to albumin. In certain embodiments, the insulinotropic peptide is linked to albumin according to any technique known to those of skill in the art. In some embodiments, the insulinotropic peptide is modified to include a reactive group which can react with available reactive functionalities on albumin to form covalent linkages.
[0255] The reactive group is chosen for its ability to form a stable covalent bond with albumin, for example, by reacting with one or more amino groups, hydroxyl groups, or thiol groups on the serum protein or peptide. Preferably, a reactive group reacts with only one amino group, hydroxyl group, or thiol group on albumin. Preferably, a reactive group reacts with a specific amino group, hydroxyl group, or thiol group on albumin. A useful conjugate of the methods described herein comprises a modified peptide, or a modified derivative thereof, which is covalently attached to albumin via a reaction of the reactive group with an amino group, hydroxyl group, or thiol group on albumin. Thus, a useful conjugate can comprise a modified peptide, or a modified derivative thereof, in which the reactive group has formed a covalent bond to albumin.
[0256] To form covalent bonds with the functional group on a protein, one may use as a chemically reactive group a wide variety of active carboxyl groups, particularly esters. While a number of different hydroxyl groups may be employed in these linking agents, the most convenient would be N-hydroxysuccinimide (NHS), N-hydroxy-sulfosuccinimide (sulfo-NHS), maleimide-benzoyl-succinimide (MBS), gamma-maleimido-butyryloxy succinimide ester (GMBS) and 3-maleimidopropionic acid (3-MPA).
[0257] Primary amines are the principal targets for NHS esters. Accessible ε-amine groups present on the N-termini of proteins react with NHS esters. However, ε -amino groups on a protein may not be desirable or available for the NHS coupling. While five amino acids have nitrogen in their side chains, only the ε -amine of lysine reacts significantly with NHS esters. An amide bond can form when the NHS ester conjugation reaction reacts with primary amines releasing N-hydroxysuccinimide. These succinimide containing reactive groups are herein referred to as succinimidyl groups.
[0258] In particular embodiments, the functional group on albumin is the single free thiol group located at amino acid residue 34 (Cys34) and the chemically reactive group is a maleimido- containing group such as (GMBA or MPA). GMBA stands for gammamaleimide-butrylamide. Such maleimide containing groups are referred to herein as maleimido groups.
[0259] In some embodiments, albumin is covalently linked to a succinimidyl or maleimido group on the insulinotropic peptide. In some embodiments, an albumin amino, hydroxyl or thiol group is covalently linked to a succinimidyl or maleimido group on the insulinitropic peptide. In some embodiments, albumin cysteine 34 thiol is covalently linked to a [2-[2-[2- maleimidopropionamido(ethoxy)ethoxy] acetic acid linker on the epsilon amino of a lysine of the insulinotropic peptide. [0260] In a specific embodiment, the reactive group is a single MPA reactive group attached to the peptide, optionally through a linking group, at a single defined amino acid and the MPA is covalently attached to albumin at substantially a single amino acid residue of albumin, preferably cysteine 34. In a preferred embodiment, the albumin is recombinant human albumin. In certain embodiments, the reactive group, preferably MPA, is attached to the peptide through one or more linking groups, preferably AEEA, AEA, or amino-octanoic acid, more particularly 8-amino- octanoic acid. In certain examples of embodiments in which the reactive group, preferably MPA, is attached to the peptide through more than one linking group, each linking group can be independently selected from the group consisting preferably of AEA ((2-amino) ethoxy acetic acid), AEEA ([2-(2-amino)ethoxy)]ethoxy acetic acid), and amino-octanoic acid, more particularly 8-amino-octanoic acid. In one embodiment, the reactive group, preferably MPA, is attached to the peptide via 1, 2, 3, 4, 5 or 6 AEEA linking groups which are arranged in tandem. In another embodiment, the reactive group, preferably MPA, is attached to the peptide via 1, 2, 3, 4, 5 or 6 8-amino-octanoic acid linking groups which are arranged in tandem.
[0261] In certain embodiments, the reactive group can be attached to any residue of the insulinotropic peptide suitable for attachment of such a reactive group. The residue can be a terminal or internal residue of the peptide. In certain embodiments, the reactive group can be attached to the carboxy-terminus or amino-terminus of the peptide. In advantageous
embodiments, the reactive group is attached to a single site of the peptide. This can be achieved using protecting groups known to those of skill in the art. In certain embodiments, a derivative of the insulinotropic peptide can comprise a residue incorporated for attachment of the reactive group. Useful residues for attachment include, but are not limited to, lysine, aspartate and glutamate residues. The residue can be incorporated internally or at a terminus of the peptide. In certain embodiments, the reactive group is attached to an internal lysine residue. In certain embodiments, the reactive group is attached to a terminal lysine residue. In certain embodiments, the reactive group is attached to an amino-terminal lysine residue. In certain embodiments, the reactive group is attached to a carboxy-terminal lysine residue, for instance, a lysine residue at the carboxy-terminus of GLP-1, GLP- 1(7-37) or exendin-4.
[0262] The manner of modifying insulinotropic peptides with a reactive group for conjugation to a macromolecule, e.g., albumin, will vary widely, depending upon the nature of the various elements comprising the insulinotropic peptide. The synthetic procedures will be selected so as to be simple, provide for high yields, and allow for a highly purified product. Normally, the chemically reactive group will be created at the last stage of insulinotropic peptide synthesis, for example, with a carboxyl group, esterification to form an active ester. Specific methods for the production of modified insulinotropic peptides are described in U.S. Patent Nos. 6,329,336, 6,849,714 or 6,887,849, the contents of each of which are incorporated by reference herein in their entireties.
[0263] The insulinotropic peptide can also be non-specifically conjugated to albumin. Bonds to amino groups will generally be employed, particularly with the formation of amide bonds for non-specific conjugation. To form such bonds, one can use as a chemically reactive group coupled to the insulinotropic peptide a wide variety of active carboxyl groups, particularly esters. While a number of different hydroxyl groups can be employed in these linking agents, the most convenient would be N-hydroxysuccinimide (NHS) and N-hydroxysulfosuccinimide (sulfo- NHS). Other linking agents which can be utilized are described in U.S. Pat. No. 5,612,034, which is hereby incorporated by reference herein in its entirety.
[0264] In some embodiments, the insulinotropic peptide conjugates can comprise an albumin fusion protein, i.e., an albumin molecule, or a fragment or variant thereof, fused to an
insulinotropic peptide. The albumin fusion protein can be generated by translation of a nucleic acid comprising a polynucleotide encoding all or a portion of a therapeutic protein joined to a polynucleotide encoding all or a portion of albumin. In some embodiments, the albumin fusion protein is comprised of albumin, or a fragment or variant thereof, fused to a glucagon-like peptide 1 as described in U.S. Patent No. 7,141,547 or 7,271,149, which are hereby incorporated by reference in their entireties. In some embodiments, the albumin fusion protein is comprised of albumin, or a fragment or variant thereof, fused to exendin-3, or a fragment or variant thereof. In some embodiments, the albumin fusion protein is comprised of albumin, or a fragment or variant thereof, fused to exendin-4, or a fragment or variant thereof. In some embodiments, the albumin fusion protein is [Gly8]GLP-l(7-36)-[Gly8]GLP-l(7-36)-human serum albumin (albiglutide) as described in U.S. Patent No. 7,141,547 or 7,271,149.
A. Insulinotropic Peptide Synthesis
[0265] Insulinotropic peptides can be synthesized by standard methods of solid phase peptide chemistry known to those of ordinary skill in the art. For example, insulinotropic peptides fragments can be synthesized by solid phase chemistry techniques following the procedures described by Steward and Young (Steward, J. M. and Young, J. D., 1984, Solid Phase Peptide Synthesis, 2nd Ed. (Pierce Chemical Company, Rockford, 111.) using an Applied Biosystem synthesizer. Similarly, multiple fragments can be synthesized then linked together to form larger fragments. These synthetic peptide fragments can also be made with amino acid substitutions at specific locations. For solid phase peptide synthesis, a summary of the many techniques may be found in J. M. Stewart and J. D. Young, 1963, Solid Phase Peptide Synthesis. (W. H. Freeman Co., San Francisco), and J. Meienhofer, 1973, Hormonal Proteins and Peptides, vol. 2, p. 46, Academic Press, New York). For classical solution synthesis see G. Schroder and K. Lupke, The Peptides, Vol. 1, (Academic Press, New York). In some embodiments, synthesis of the insulinotropic peptides is as described in U.S. Patent Nos. 6, 329,336, 6,849,714 or 6,887,849, the contents of each of which are incorporated by reference herein in their entireties.
B. Conjugation
[0266] Preferably, the peptide and albumin are present in the conjugate in a 1: 1 molar ratio, or an approximately 1 : 1 molar ratio. In a preferred embodiment, the peptide and albumin are present in the conjugate in a 1 : 1 molar ratio, or an approximately 1 : 1 molar ratio, and the peptide is attached to the reactive group, optionally through a linking group, at substantially only one site on the peptide and the reactive group is attached to the albumin at substantially only one site on albumin.
[0267] Preferably, the albumin in the peptide conjugates is human serum albumin. Preferably, the single site of attachment of the reactive group to albumin is preferably the thiol of cysteine 34 of albumin (e.g., via a maleimide linkage). In a specific embodiment, the reactive group is a single MPA reactive group attached to the peptide, optionally through a linking group, at a single defined amino acid and the MPA is covalently attached to albumin at substantially a single amino acid residue of albumin, preferably cysteine 34.
[0268] In a preferred embodiment, a conjugate is formed by contacting a modified peptide comprising a maleimido group with a thiol-containing serum protein, preferably albumin, under conditions comprising a pH of between 3.0 and 8.0, thereby preferably forming a stable thioether linkage which cannot be cleaved under physiological conditions. In preferred embodiments, the serum protein is recombinant human albumin.
[0269] In one embodiment, the modified peptide of the conjugate is amidated at its C-terminal end. In another embodiment, the modified peptide is not amidated at its C-terminal end. A conjugate can also comprise such an amidated peptide.
[0270] In a preferred embodiment, a single reactive group is covalently attached at a defined site of the modified peptide. In a preferred embodiment of the conjugate, a single reactive group is covalently attached at a defined site of the modified peptide and the reactive group is covalently attached to a single defined site of albumin, preferably to the thiol group of amino acid residue Cys34 of albumin. Preferably, the reactive group of a modified peptide or conjugate of the invention comprises a maleimide group and forms peptide: albumin conjugates of
approximately a 1 : 1 molar ratio. In certain embodiments, a 1 : 1 molar ratio of peptide to serum protein is preferred over higher ratios because a 1 : 1 molar ratio provides better biological activity and less immunogenicity than higher ratios (see e.g., Stehle et al. 1997 Anti-Cancer Drugs 8:677- 685, incorporated by reference herein in its entirety). [0271] In a preferred embodiment, the albumin is recombinant human albumin. Specific methods for the production of preformed peptide: albumin conjugates are described in U.S.
Provisional Application No. 60/791,241, entitled " Process for the Production of Preformed Conjugate of Recombinant Albumin," filed April 11, 2006, and U.S. Pat. App. No. 11/645,297 (Publication No. 2007/0269863), entitled "Process for the Production of Preformed Conjugates of Albumin and a Therapeutic Agent," filed December 22, 2006, the contents of each of which are incorporated by reference herein in their entireties. Specific methods for the purification of peptide: albumin conjugates are described in U.S. Patent Application Publication No.
2005/0267293, which is incorporated by reference herein in its entirety.
[0272] In certain embodiments, the conjugate is according to the following:
Figure imgf000072_0001
(SEQ ID NO: 31) wherein X is S, O, or NH of an amino acid of said protein. In certain embodiments, said protein is albumin. In certain embodiments, said protein is albumin and X is S (sulfur) of Cys 34 of said albumin. Albumin of the conjugate can be any albumin as described above.
[0273] In certain embodiments, the conjugate is according to the following:
Figure imgf000072_0002
(SEQ ID NO: 32) wherein X is S, O, or NH of an amino acid of said protein. In certain embodiments, said protein is albumin. In certain embodiments, said protein is albumin and X is S (sulfur) of Cys 34 of said albumin. The albumin of the conjugate can be any albumin as described below.
a. Albumin
[0274] Any albumin known to those of skill in the art can be used to form a insulinotropic peptide albumin conjugate of the formulations described herein. In some embodiments, the albumin can be serum albumin isolated from a host species and purified for use in the formation of a conjugate. The serum albumin can be any mammalian serum albumin known to those of skill in the art, including but not limited to mouse, rat, rabbit, guinea pig, dog, cat, sheep, bovine, ovine, equine, or human albumin. In some embodiments, the albumin is human serum albumin. In some embodiments, the albumin is bovine serum albumin.
[0275] Human serum albumin (HSA) is responsible for a significant proportion of the osmotic pressure of serum and also functions as a carrier of endogenous and exogenous ligands. In its mature form, HSA is a non-glycosylated monomeric protein of 585 amino acids, corresponding to a molecular weight of about 66 kD. Its globular structure is maintained by 17 disulfide bridges which create a sequential series of 9 double loops. See Brown, J.R., Albumin Structure, Function and Uses, Rosenoer, V.M. et a/.(eds), Pergamon Press, Oxford (1977), which is incorporated by reference herein in its entirety. The native mature human serum albumin sequence is:
DAHKSE VAHRFKDLGE ENFKALVLIA FAQYLQQCPF EDHVKLVNEV TEFAKTCVAD ESAENCDKSL HTLFGDKLCT VATLRETYGE MADCCAKQEP ERNECFLQHK DDNPNLPRLV RPEVDVMCTA FHDNEETFLK KYLYEIARRH PYFYAPELLF FAKRYKAAFT ECCQAADKAA CLLPKLDELR DEGKASSAKQ RLKCASLQKF GERAFKAWAV ARLSQRFPKA EFAEVSKLVT DLTKVHTECC HGDLLECADD RADLAKYICE NQDSISSKLK ECCEKPLLEK SHCIAEVEND EMPADLPSLA ADFVESKDVC KNYAEAKDVF LGMFLYEYAR RHPDYSVVLL LRLAKTYETT LEKCCAAADP HECYAKVFDE FKPLVEEPQN LIKQNCELFE QLGEYKFQNA LLVRYTKKVP QVSTPTLVEV SRNLGKVGSK CCKHPEAKRM PCAEDYLSVV LNQLCVLHEK TPVSDRVTKC CTESLVNRRP CFSALEVDET YVPKEFNAET FTFHADICTL SEKERQIKKQ TALVELVKHK PKATKEQLKA VMDDFAAFVE KCCKADDKET CFAEEGKKLV AASQAALGL (SEQ ID NO. 30).
[0276] Thus, conjugates formed with the mature form of albumin are within the scope of the processes described herein. Unless indicated otherwise, reference to an albumin herein is intended to refer to the mature form of the albumin.
[0277] In some embodiments, the albumin is recombinant albumin. The recombinant albumin can be any mammalian albumin known to those of skill in the art, including but not limited to mouse, rat, rabbit, guinea pig, dog, cat, sheep, bovine, ovine, equine, or human albumin. In a preferred embodiment, the recombinant albumin is recombinant human albumin (rHA). In some embodiments, the recombinant human albumin is recombinant human serum albumin. In various embodiments, rHA can be produced in a mammalian or non-mammalian organism. In one embodiment, the rHA is produced in a non-mammalian organism. Examples of non-mammalian organisms that can be used for the production of rHA include, without limitation, yeast, bacteria, plants, fungi, and insects. In one embodiment, the rHA is produced in a whole plant or a whole fungus. In another embodiment, the rHA is produced in cultured plant cells, cultured fungus cells, or cultured insect cells. In another embodiment, the rHA is produced in a non-human mammal or in non-human mammalian cells. Examples of non-human mammals that can be used for the production of rHA include, without limitation, those belonging to one of the following: the family Bovidae, the family Canidae, the family Suidae, the order Rodentia, the order Lagomorpha, and the order Primates (excluding humans). In a particular embodiment, the non-human mammal that is used for the production of rHA is selected from the group consisting of a cow, a dog, a pig, a sheep, a goat, a rat, a mouse, a rabbit, a chimpanzee, and a gorilla. In another embodiment, the non-human mammalian cells used for the production of rHA are, without limitation, bovine, canine, porcine, ovine, caprine, rodent, rabbit, or non-human primate cells. The main advantage of rHA produced in a non-human organism compared with albumin purified from human blood or serous fluids is the absence of human-derived products in the manufacturing process of rHA. The use of such controlled production methods leads to a purer product with less structural heterogeneity.
[0278] In some embodiments, the insulinotropic peptide albumin conjugate can comprise an albumin precursor. Human albumin is synthesized in liver hepatocytes and then secreted in the blood stream. This synthesis leads, in a first instance, to a precursor, prepro-HSA, which comprises a signal sequence of 18 amino acids directing the nascent polypeptide into the secretory pathway. Thus, conjugates formed with an albumin precursor are within the scope of the conjugates described herein.
[0279] In certain embodiments, the insulinotropic peptide albumin conjugate can comprise molecular variants of albumin. Variants of albumin can include natural variants resulting from the polymorphism of albumin in the human population. More than 30 apparently different genetic variants of human serum albumin have been identified by electrophoretic analysis under various conditions. See e.g., Weitkamp et ai, Ann. Hum. Genet., 36(4):381-92 (1973); Weitkamp, Isr. J. Med. Set, 9(9):1238-48 (1973); Fine et ai, Biomedicine, 25(8):291-4 (1976); Fine et ai, Rev. Fr. Transfus. ImmunohematoL, 25(2): 149-63. (1982); Rochu et ai, Rev. Fr. Transfus.
Immunohematol. 31(5):725-33 (1988); Arai et ai, Proc. Natl. Acad. Sci. U.S.A 86(2): 434-8 (1989), the contents of each of which are incorporated by reference herein in their entireties. Thus, conjugates formed with molecular variants of albumin are within the scope of the conjugates described herein.
[0280] In certain embodiments, the insulinotropic peptide albumin conjugate can comprise mercaptalbumin enriched albumin. While not intending to be bound by any particular theory of operation, it is believed that oxidation, or "capping" of the cysteine 34 thiol of albumin by cysteine, glutathione, metal ions, or other adducts can reduce the specificity of conjugation to the reactive group of the compound. Accordingly, mercaptalbumin can be enriched from heterogeneous pools of reduced and oxidized albumin by contact with agents known in the art to be capable of converting capped albumin-Cys34 to albumin-Cys34-SH. In certain embodiments, the mercaptalbumin can be enriched by contacting the albumin with thioglycolic acid (TGA). In certain embodiments, the mercaptalbumin can be enriched by contacting the albumin with dithiothreitol (DTT). In some embodiments, mercaptalbumin may be enriched by subjecting the albumin to hydrophobic interaction chromatography, using phenyl or butyl sepharose, or a combination thereof. In other embodiments, mercaptalbumin may be enriched by contacting the albumin with TGA or DTT, followed by purification by hydrophobic interaction chromatography, using phenyl or butyl sepharose resin, or both.
[0281] In a specific embodiment, the albumin variant has not more than 5, 4, 3, 2 or 1 amino acid substitutions, deletions or additions relative to the sequence of mature native human serum albumin.
[0282] In some embodiments, the insulinotropic peptide albumin conjugate can comprise derivatives of albumin which share substantial homology with albumin. For instance, conjugates can be formed with an albumin homologue having an amino acid sequence which shares at least 75%, at least 80%, at least 85%, more preferably at least 90%, and most preferably at least 95% identity to native human serum albumin, i.e., SEQ ID NO. 30. Percent identity in this context means the percentage of amino acid residues in the candidate sequence that are identical (i.e., the amino acid residues at a given position in the alignment are the same residue) or similar (i.e., the amino acid substitution at a given position in the alignment is a conservative substitution, as discussed above), to the corresponding amino acid residue in the peptide after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence homology. In certain embodiments, an albumin derivative is characterized by its percent sequence identity or percent sequence similarity with the naturally occurring albumin sequence. Sequence homology, including percentages of sequence identity and similarity, are determined using sequence alignment techniques well-known in the art, preferably computer algorithms designed for this purpose, such as BLAST, using the default parameters of said computer algorithms or the software packages containing them.
[0283] In certain embodiments, the albumin homologue comprises a free cysteine. In certain embodiments, the albumin homologue comprises a single free cysteine. In some embodiments, the albumin homologue comprises a free cysteine 34.
[0284] In some embodiments, the insulinotropic peptide albumin conjugate can comprise an N- terminal fragment of human serum albumin of at least 100, 200, 300, 400, 500 or more than 500 amino acids. In another embodiment, the insulinotropic peptide albumin conjugate can comprise a human serum albumin variant comprising a modification of the Asp-Ala-His-Lys N-terminal sequence. In another embodiment, the insulinotropic peptide albumin conjugate can comprise at least one deletion among the three N-terminal amino acid residues Asp-Ala-His. In another embodiment, the insulinotropic peptide albumin conjugate can comprise an N- terminal extension of albumin, such as Glu 3, Ala"2, Glu 1, Phe°-HSA (1-585 of SEQ ID NO. 30) or an N-terminal fragment thereof. In another embodiment of the invention the human serum albumin (HSA) variant is selected from the group consisting of HSA (2-585 of SEQ ID NO. 30), HSA (3-585 of SEQ ID NO. 30), HSA (4-585 of SEQ ID NO. 30), Asp-Ala- HSA (4-585 of SEQ ID NO. 30), Xaa3-HSA (1-585 of SEQ ID NO. 30) where Xaa3 is an amino acid residue which has substituted the His residue occupying position 3 in native HSA, and N-terminal fragments thereof.
[0285] In some embodiments, the insulinotropic peptide albumin conjugate can comprise structural derivatives of albumin. Structural derivatives of albumin can include proteins or peptides which possess an albumin-type activity, for example, a functional fragment of albumin. In some embodiments, the derivative is an antigenic determinant of albumin, i.e., a portion of a polypeptide that can be recognized by an anti-albumin antibody. In some embodiments, the recombinant albumin can be any protein with preferably a plasma half-life of 75% to 100% of the plasma half-life of human serum albumin in humans and which can be obtained by modification of a gene encoding human serum albumin. By way of example and not limitation, the
recombinant albumin can contain additions or deletions in the trace metal binding region of albumin, such that binding of trace metals, e.g., nickel and/or copper is reduced or eliminated, as described in U.S. Patent No. 6,787,636, which is incorporated by reference herein in its entirety. In particular, the recombinant albumin can be modified in the N-terminal region or binding region VI, such as through a truncation of at least one amino acid at the N-terminal end, so that it exhibits reduced or eliminated binding of trace metals such as nickel and/or copper. Other suitable modifications to this binding region include mutations such as an elongation or insertion which will be sufficient to disrupt the trace metal binding which is highest at this site. Reduced trace metal binding by albumin can be advantageous for reducing the likelihood of an allergic reaction to the trace metal in the subject being treated with the albumin composition.
[0286] Structural derivatives of albumin can be generated using any method known to those of skill in the art, including but not limited to, oligonucleotide-mediated (site-directed) mutagenesis, alanine scanning, and polymerase chain reaction (PCR) mutagenesis. Site -directed mutagenesis (see Carter, Biochem. J. 237: 1-7 (1986); Zoller and Smith, Methods Enzymol. 154:329-50 (1987)), cassette mutagenesis, restriction selection mutagenesis (Wells et ai, Gene 34:315-323 (1985)) or other known techniques can be performed on cloned albumin-encoding DNA to produce albumin variant DNA or sequences which encode structural derivatives of albumin (Ausubel et ai, Current Protocols In Molecular Biology, John Wiley and Sons, New York (current edition); Sambrook et ah, Molecular Cloning, A Laboratory Manual, 3d. ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (2001), the contents of each of which are incorporated by reference herein in their entireties.
[0287] In certain embodiments, albumin derivatives include any macromolecule with preferably a plasma half-life of 75% to 100% of the plasma half-life of human serum albumin in humans which can be obtained by in vitro modification of the albumin protein. In some embodiments, the albumin is modified with fatty acids. In some embodiments, the albumin is modified with metal ions. In some embodiments, the albumin is modified with small molecules having high affinity to albumin. In some embodiments, the albumin is modified with sugars, including but not limited to, glucose, lactose, mannose, and galactose.
[0288] In some embodiments, the insulinotropic peptide albumin conjugate can comprise an albumin fusion protein, i.e., an albumin molecule, or a fragment or variant thereof, fused to a therapeutic protein, or a fragment or variant thereof. The albumin fusion protein can be generated by translation of a nucleic acid comprising a polynucleotide encoding all or a portion of a therapeutic protein joined to a polynucleotide encoding all or a portion of albumin. Any albumin fusion protein known to those of skill in the art can be used to form conjugates according to the processes of the invention. Exemplary albumin fusion proteins are described in U.S. Patent Nos. 6,548,653, 6,686,179, 6,905,688, 6,994,857, 7,045,318, 7,056,701, 7,141,547 and 7,271,149, the contents of each of which are incorporated by reference herein in their entireties. In some embodiments, the albumin fusion protein is comprised of albumin, or a fragment or variant thereof, fused to a glucagon-like peptide 1 as described in U.S. Patent No. 7,141,547 or
7,271,149. In some embodiments, the albumin fusion protein is comprised of albumin, or a fragment or variant thereof, fused to exendin-3, or a fragment or variant thereof. In some embodiments, the albumin fusion protein is comprised of albumin, or a fragment or variant thereof, fused to exendin-4, or a fragment or variant thereof. In some embodiments, the albumin fusion protein is comprised of albumin, or a fragment or variant thereof, fused to a multiyear of exendin-4, or a fragment or variant thereof.
[0289] Albumin used to form a conjugate described herein can be obtained using methods or materials known to those of skill in the art. For instance, albumin can be obtained from a commercial source, e.g., Novozymes Biopharma UK Ltd. (Nottingham, UK; recombinant human albumin produced by Saccharomyces cerevisiae); Cortex-Biochem (San Leandro, Calif.; serum albumin), Talecris Biotherapeutics (Research Triangle Park, North Carolina; serum albumin), ZLB Behring (King of Prussia, PA), or New Century Pharmaceuticals (Huntsville, Ala.;
recombinant human albumin derived from Pichia pastoris). [0290] In some embodiments, the albumin derivative is RECOMBUMIN® (Novozymes Biopharma UK Ltd. (Nottingham, UK)). Recombumin® is a recombinant human albumin (rHA) that is produced in vitro using recombinant yeast technology, in which genetically modified yeast (Saccharomyces cerevisiae) secrete soluble rHA which is subsequently harvested, purified and formulated for use as an excipient for the manufacture of biologies or a coating for medical devices. The main advantage of rHA over HSA is that it is expressed in yeast with no animal- or human-derived products used in the manufacturing process. The use of such controlled production methods leads to a purer product with less structural heterogeneity. Previous studies have indicated that there is no significant difference between soluble rHA and HSA in terms of their biochemical characteristics, radiolabelling efficiency and biological behavior in vitro and in vivo. See Dodsworth et ah, 1996, Biotechnol. Appl. Biochem. 24: 171-176.
[0291] In some embodiments, the albumin derivative is MEDWAY® (ALBREC®, GB-1057, Mitsubishi Tanabe Pharma Corp., Osaka, Japan). MEDWAY is a recombinant human albumin (rHA) that is produced in vitro using recombinant yeast technology, in which genetically modified yeast (Pichia pastoris) secrete soluble rHA which can be subsequently harvested, purified and formulated for the indicated treatment.
[0292] In some embodiments, the albumin derivative is ALBAGEN™ (New Century Pharma, Huntsville, AL). ALBAGEN is HSA (2-585) and is hypoallergenic due to the modified metal binding properties caused by the single N-terminal deletion.
[0293] In some embodiments, the albumin derivative is ALBUCULT™ (Novozymes
Biopharma UK Ltd. (Nottingham, UK)). Albucult™ is a yeast-derived recombinant human albumin solution designed specifically for cell culture applications. It is produced without the use of animal- or human-derived materials and is therefore free from risk of contaminating human or animal-derived viruses or prions.
VI. Pharmaceutical Formulations
[0294] In the methods provided herein, the insulinotropic peptide or modified insulinotropic peptide can be administered in a pharmaceutical formulation deemed suitable by one of skill in the art. The formulation can be suitable for administration via a parenteral route such as subcutaneous, intravenous, intramuscular, transdermal, intra-arterial, or intraperitoneal routes, or via other routes such as oral, topical, or inhalation routes.
[0295] In some embodiments, the concentration of the insulinotropic peptide or modified insulinotropic peptide in the formulation is from about 0.1 mg/ml to about 100 mg/ml, from about 0.1 mg/ml to about 75 mg/ml, from about 0.1 mg/ml to about 50 mg/ml, from about 0.1 mg/ml to about 40 mg/ml, from about 0.1 mg/ml to about 30 mg/ml, from about 1 mg/ml to about 100 mg/ml, from about 5 mg/ml to about 50 mg/ml, or from about 10 mg/ml to 20 mg/ml. In some embodiments, the concentration of the insulinotropic peptide or modified insulinotropic peptide in the formulation is higher than about 10 mg/ml, about 20 mg/ml, about 50 mg/ml, about 100 mg/ml, about 200 mg/ml, or about 500 mg/ml. In some embodiments, the concentration of the insulinotropic peptide or modified insulinotropic peptide in the formulation is lower than about 100 mg/ml, about 50 mg/ml, about 40 mg/ml, about 30 mg/ml, about 20 mg/ml, about 10 mg/ml, about 5 mg/ml, about 1 mg/ml, or about 0.1 mg/ml. In preferred embodiments, the concentration of the insulinotropic peptide or modified insulinotropic peptide in the formulation is about 1 mg/ml to about 50 mg/ml, from about 1 mg/ml to about 40 mg/ml, from about 1 mg/ml to about 20 mg/ml, or from about 1 to about 15 mg/ml. In particularly preferred embodiments, the concentration of the insulinotropic peptide or modified insulinotropic peptide in the formulation is about 1 mg/ml. In other particularly preferred embodiments, the concentration of the insulinotropic peptide or modified insulinotropic peptide in the formulation is about 2.5 mg/ml. In other particularly preferred embodiments, the concentration of the insulinotropic peptide or modified insulinotropic peptide in the formulation is about 5 mg/ml. In other particularly preferred embodiments, the concentration of the insulinotropic peptide or modified insulinotropic peptide in the formulation is about 10 mg/ml. In some embodiments of any of the above concentrations, the amount of the the insulinotropic peptide or modified insulinotropic peptide formulation is about 1 ml. In some embodiments, the administration is subcutaneous.
[0296] In certain embodiments, the formulations herein can be administered as monotherapy. In other words, the formulations herein can be provided as the sole administration of an active agent for treatment of one or more conditions provided herein.
[0297] The formulations described herein can also be administered in combination with or can comprise one or more second therapeutic agents useful for the particular indication being treated, preferably those with complementary activities that do not adversely affect the insulinotropic peptide conjugate of the formulation. In certain embodiments, such second therapeutic agents can be present with the insulinotropic peptide conjugate in amounts that are effective for the purpose intended. In a particular embodiment, the second therapeutic agent is an anti-diabetic agent, e.g., an oral anti-diabetic agent, e.g., a biguanide, e.g., metformin.
[0298] In some embodiments, the pharmaceutical formulations comprise a buffer that maintains a physiologically suitable pH. Exemplary physiologically suitable pH ranges are provided below. In addition, the buffer can serve to enhance isotonicity and chemical stability of the formulation. In other embodiments, the pharmaceutical formulations do not comprise a buffer.
[0299] In some embodiments, the formulation has a pH of about 3.0 to 8.0. In some embodiments, the formulation has a pH of about 4.0 to 8.0. In some embodiments, the formulation has a pH of about 4.5 to 7.0. In some embodiments, the formulation has a pH of about 4.0 to 6.0. In some embodiments, the formulation has a pH of about 6.0 to 8.0. In some embodiments, the formulation has a pH of about 6.0 to 9.0. In some embodiments, the formulation has a pH of about 6.5 to 8.0. In some embodiments, the formulation has a pH of about 5.0 to 7.0. In some embodiments, the formulation has a pH of about 4.5 to 6.0. In some embodiments, the formulation has a pH of about 5.0 to 6.0. In some embodiments, the formulation has a pH of about 5.5 to 6.5. In some embodiments, the formulation has a pH of about 6.5 to 7.5. In some embodiments, the formulation has a pH of about 5.1 to 6.0, about 5.2 to 6.0, about 5.3 to 6.0, about 5.4 to 6.0, about 5.5 to 6.0, about 5.6 to 6.0, about 5.7 to 6.0, or about 5.8 to 6.0. In some embodiments, said formulation has a pH of about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9 or 9.0. In a particular embodiment, the formulation has a pH of about 5.0. In a particular embodiment, the formulation has a pH of about 5.5. In another particular embodiment, the formulation has a pH of about 6.0. In a particular embodiment, the formulation has a pH of about 6.5. In another particular embodiment, the formulation has a pH of about 7.0. In another particular embodiment, the formulation has a pH of about 7.5. The pH can be adjusted as necessary by techniques known in the art. For example, hydrochloric acid or sodium hydroxide can be added as necessary to adjust the pH to desired levels.
[0300] Useful buffers in the buffered formulations described herein include, but are not limited to, acetate, phosphate, maleate, succinate, histidine (e.g., L-histidine),
tris(tris(hydroxymethyl)aminomethane), diethanolamine, citrate, other organic acids and mixtures thereof, e.g., citrate-phosphate. The formulation can further comprise any counter-ion deemed suitable, such as sodium or calcium.
[0301] In some embodiments, the buffer is present in an amount sufficient to maintain suitable pH. In some embodiments, the buffer is present in the formulations from about 0.1 mM to about 100 mM, from about 0.1 mM to about 50 mM, from about 0.1 mM to about 30 mM, about 0.1 mM to about 25 mM, from about 0.1 mM to about 20 mM, or from about 5 mM to about 15 mM. In certain embodiments, the buffer is at about 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, or 15 mM. In some embodiments, the buffer is at about 20 mM, 25 mM, 30 mM, 35 mM, or 40 mM. In some embodiments, the buffer is a sodium acetate buffer or a sodium phosphate buffer at about 10 mM.
[0302] The formulations can comprise a tonicity modifier that contributes to maintain the isotonicity of the formulation. In some embodiments, the formulation is isotonic, i.e., the formulation possesses the same or about the same osmotic pressure as blood plasma. Isotonic formulations will generally have an osmotic pressure from about 250 to 350 mOsm, prefereably from about 250 to about 330 mOsm. In some embodiments, the formulation is hypertonic. In some embodiments, the formulation is hypotonic.
[0303] The tonicity modifier can be any tonicity modifier apparent to one of skill, such as a salt, a sugar, a sugar alcohol, a polyol or an amino acid. Exemplary tonicity modifiers include but are not limited to a salt such as sodium chloride, calcium chloride or magnesium chloride, a sugar or polyol such as lactose, sorbitol, sucrose, mannitol, trehalose, raffinose, polyethylene glycol, hydroxyethyl starch, glycine and combinations thereof. In some preferred embodiments, the tonicity modifier is sodium chloride. In other preferred embodiments, the tonicity modifier is sorbitol. In certain embodiments, combined tonicity modifiers yield a total osmolality that is isotonic as described above.
[0304] When the formulation is a lyophilized formulation, salts or non-reducing sugars are preferred as tonicity modifiers. A " non-reducing sugar" is one which does not contain a hemiacetal group that can reduce metal ions or react covalently with lysine and other amino groups in proteins. Non-reducing sugars include sucrose, trehalose, sorbose, melezitose and raffinose. Non-reducing sugars can prevent or reduce chemical and/or physical instability of the peptides upon lyophilization and subsequent storage.
[0305] The tonicity modifier is present in the formulation in an amount to maintain desired tonicity of the formulation. In some embodiments, the tonicity modifier is present at about 0.1 % to about 50 % (w/v), about 0.5 % to about 20 % (w/v), about 1 % to about 10 % (w/v), or about 4 % to about 6 % (w/v). In some embodiments, the tonicity modifier is present at about 5 % (w/v). In some embodiments, the tonicity modifier is present at a concentration of at least 1 mM. In some embodiments, the tonicity modifier is present at about 1 mM to about 200 mM, from about 10 mM to about 150 mM or from about 50 mM to about 100 mM. In some preferred
embodiments, the formulation comprises about 135 mM sodium chloride. In other preferred embodiments, the formulation comprises about 150 mM sodium chloride. In other preferred embodiments, the formulation comprises about 5% sorbitol (w/v).
[0306] The formulations can also comprise a stabilizer to stabilize the conjugate during fluctuations in storage temperature and to minimize degradation products, peptide degradants and aggregation. Useful stabilizers in the formulations of the invention include, but are not limited to, sodium octanoate, myristic acid, Na-N-acetyltryptophan, L-glutamic acid, arginine, nitrogen and combinations thereof. In particular embodiments, the stabilizer is sodium octanoate. In particular embodiments, the stabilizer is myristic acid.
[0307] In certain embodiments, the stabilizer is present in the formulation at about 0.1 mM to 30 mM, about 0.5 mM and 20 mM, about 1 mM to about 15 mM, or about 5 mM to about 10 mM. In certain embodiments, the stabilizer is present in the formulation at about 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, 15 mM, 16 mM, 17 mM, 18 mM, 19 mM or 20 mM. In a particular embodiment, the stabilizer is sodium octanoate at about 5 mM. In another particular embodiment, the stabilizer is sodium octanoate at about 15 mM. In another particular embodiment, the stabilizer is myristic acid at about 0.1 mM.
[0308] The formulations can also comprise a surfactant. Surfactants are compounds that reduce interfacial tension between a liquid and a solid when dissolved in solution, and can be added to the formulation to reduce aggregation of the reconstituted protein and/or reduce the formation of particulates in the reconstituted formulation. Exemplary surfactants include polysorbates (e.g. polysorbates 20 or 80); poloxamers (e.g. poloxamer 188 (pluronic F68)); Triton; sodium dodecyl sulfate (SDS); sodium laurel sulfate; sodium octyl glycoside; lauryl-, myristyl-, linoleyl-, or stearyl-sulfobetaine; lauryl-, myristyl-, linoleyl- or stearylsarcosine; linoleyl-, myristyl-, or cetyl- betaine; lauroamidopropyl-, cocamidopropyl-, linoleamidopropyl-, myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-betaine (e.g. lauroamidopropyl); myristamidopropyl-, palmidopropyl-, or isostearamidopropyldimethylamine; sodium methyl cocoyl-, or disodium methyl oleyl-taurate; and the MONAQUAT™ series (Mona Industries, Inc., Paterson, N.J.), polyethyl glycol, polypropyl glycol, and copolymers of ethylene and propylene glycol, etc.
[0309] The amount of surfactant is such that it reduces aggregation of the formulated peptides or peptide conjugates and/or minimizes the formation of particulates in the formulation and/or reduces adsorption. For example, the surfactant can be present in the formulation in an amount of about 0.001-1 % (w/v), and preferably, about 0.01-0.5% (w/v). In some embodiments, the formulation comprises a surfactant which is a poloxamer. In some embodiments, the formulation comprises pluronic F68. In particular embodiments, the formulation comprises between about 0.01% (w/v) and about 1% (w/v) pluronic F68, more preferably about 0.1% (w/v) pluronic F68.
[0310] In certain embodiments, the formulations comprise the above-identified agents (i.e. insulinotropic peptide conjugates, buffer, tonicity modifier, stabilizer and surfactant) and are free of one or more preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and benzethonium chloride. In other embodiments, a preservative can be included in the formulations, particularly where the formulations are multi-use formulations. Exemplary preservatives include but are not limited to m-cresol, benzyl alcohol, methanol, ethanol, isopropanol, butyl paraben, ethyl paraben, methyl paraben, phenol, glycerol, xylitol, resorcinol, cathechol, 2, 6- dimethylcyclohexanol, 2-methyl-2,4-pentadiol, dextran, polyvinylpyrrolidone, 2-chlorophenol, benzethonium chloride, merthiolate (thimerosal), benzoic acid (propyl paraben) MW 180.2, benzoic acid MW 122.12, benzalkonium chloride, chlorobutanol, sodium benzoate, sodium propionate, and cetylpyridinium chloride. In certain embodiments, the preservatives are mercury- free. Any of these preservatives can be used as a sole preservative or in combination with each other in the presently disclosed formulations.
[0311] In preferred embodiments, preservatives that are compatible with the buffer and other components of the formulations (i.e., the solution is clear) are used. When the buffer is sodium acetate or sodium phosphate, compatible preservatives include methanol, ethanol, iso-propanol, glycerol, resorcinol, 2-methyl-2,4-pentadiol, merthiolate (thimerosal), benzalkonium chloride, sodium benzoate, cetylpyridinium chloride.
[0312] The concentration of the preservative used in the formulations can be determined according to the judgment of those of skill in the art. In some embodiments, about 0.005 to 10 % (w/v), about 0.1 to 1.0 % (w/v), or about 0.3 to 0.7 % (w/v) of the preservative is present in the formulations. In some embodiments, about 0.005, 0.1, 0.3, 0.5, 0.7, or 1.0 % (w/v) of the preservative is present in the formulations.
[0313] A bulking agent can be included in a lyophilized formulation to facilitate the production of an essentially uniform lyophilized cake which maintains an open pore structure. Exemplary bulking agents include mannitol, glycine, polyethylene glycol and xorbitol. Bulking agents can also serve as a tonicity modifier as well.
[0314] One or more other pharmaceutically acceptable carriers, excipients or stabilizers, for example, such as described in Remington's Pharmaceutical Sciences 19th edition, Genarro, A. Ed. (1995) can be included in the formulations provided that they do not significantly adversely affect the desired characteristics of the formulation. Additional constituent elements of the formulations of the present invention can include water, e.g., water for injection, vegetable oil, a thickening agent such as methylcellulose antiadsorbant, a wetting agent, antioxidants including ascorbic acid and methionine, chelating agents such as EDTA, metal complexes (e.g. Zn-protein complexes), biodegradable polymers such as polyesters, and/or salt-forming counterions such as sodium etc. Acceptable carriers, excipients or stabilizers are present in an amount such that they are nontoxic to subjects at the dosages and concentrations employed.
[0315] The optimal formulation according to the present invention can vary depending on factors such as the amount of time the formulation will be stored, conditions under which the formulation will be stored and used, the particular subject population to which the formulation may be administered, etc. Preferably, the formulation is stable at room temperature (about 25 °C) or at 40 °C for at least 1, 2, 3, 4, 5, 6, 9, 12, 18, 24 or 36 months and/or stable at about 2-8 °C for at least 1, 2, 3, 4, 5, 6, 9, 12, 18, 24 or 36 months. Furthermore, in certain embodiments, the formulation is preferably stable following freezing (e.g., -70 °C). In certain embodiments, the criteria for stability are as follows: (1) the formulation remains clear by visual analysis; (2) the concentration, pH and osmolality of the formulation has no more than about ± 10% change; (3) no more than about 10%, more preferably no more than about 5%, or most preferably no more than about 1% of aggregate forms as measured by SEC-HPLC; and (4) no more than 10%, more preferably no more than about 5%, or most preferably no more than 1% of peptide or peptide conjugate breaks down as measured by SDS-PAGE or RP-HPLC.
[0316] In certain embodiments, the formulations as described herein can be contained in a vial, bottle, tube, syringe or other container for single or multiple administrations. Such containers can be made of glass or a polymer material such as polypropylene, polyethylene, polyvinylchloride, or polyolefin, for example. In some embodiments, the containers can include a seal, or other closure system, such as a rubber stopper that can be penetrated by a needle in order to withdraw a single dose and then re-seal upon removal of the needle. All such containers for injectable liquids, lyophilized formulations, reconstituted lyophilized formulations or reconstitutable powders for injection known in the art are contemplated for use in the presently disclosed methods and formulations. In certain embodiments, the insulinotropic peptide or modified insulinotropic peptide is provided as a lyophilized formulation and reconstituted prior administration. In particular embodiments, the lyophilized insulinotropic peptide or modified insulinotropic peptide is provided in a 2-chamber syringe and reconstituted in the syringe. In particular embodiments, the lyophilized insulinotropic peptide or modified insulinotropic peptide is provided in a 2- chamber cartridge and reconstituted in the cartridge. In a particular embodiment, the container is a pen-type delivery apparatus comprising a single dose or multiple doses. Such a pen-type delivery apparatus can be permanent, e.g., a permanent pen that houses a disposable cartridge containing a single dose or multiple doses, or the entire apparatus can be disposable, e.g., a disposable pen that contains a single dose or multiple doses. In certain embodiments where the pen-type delivery apparatus comprises multiple doses, the dose can be pre-set, i.e., fixed. In other embodiments, the dose can be a flexible dose, i.e., dialed-in by the user. In some embodiments, the pen-type delivery apparatus comprises a luer-lock, luer-cone, or other needle fitting connector that facilitates attachment of a disposable needle. In other embodiments, the pen-type delivery apparatus comprises a staked, i.e., permanent needle. In another particular embodiment, the container is a syringe. In some embodiments, the syringe comprises a luer-lock, luer-cone, or other needle fitting connector that facilitates attachment of a disposable needle. In other embodiments, the syringe comprises a staked, i.e., permanent, needle. In some embodiments, the syringe is prefilled with a single dose or multiple doses.
[0317] The formulations provided herein can be formulated in a variety of concentrations in various vial and cartridge sizes for various administration dosages. For example, the dosages can be formulated in a 0.25, 0.5, 1, 2 or 3 ml vial, or any other size vial or other container known by one of skill in the art. [0318] The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to, or following, preparation of the formulation. Alternatively, sterility of the entire formulation can be accomplished by autoclaving the ingredients, except for protein, at about 120°C for about 30 minutes, for example.
[0319] In certain embodiments, the present invention provides a pharmaceutical formulation comprising a conjugate of albumin to exendin-4, or a derivative thereof, at a concentration from about 1 mg/ml to about 100 mg/ml, optionally a buffer, a tonicity modifier, a stabilizer, a surfactant and optionally a preservative, wherein said formulation has a pH from about 4 to about 8.
[0320] In certain embodiments, the pharmaceutical formulation comprises, or alternatively consists of, a conjugate of albumin and an insulinotropic peptide, said insulinotropic peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence, said conjugate being at a concentration of about 1 mg/ml to about 100 mg/ml; optionally a buffer; a tonicity modifier, wherein the tonicity modifier is at a concentration of at least 1 mM; a stabilizer; a surfactant, and optionally a preservative, wherein said formulation has a pH from about 4 to about 8.
[0321] In certain embodiments, the exendin-4 albumin conjugate comprises recombinant human serum albumin cysteine 34 thiol covalently linked to a [2- [2- [2
maleimidopropionamido(ethoxy)ethoxy] acetic acid linker on the epsilon amino of the carboxy terminal lysine of exendin-4(l-39)Lys40-NH2. Such a conjugate can be formed by covalently bonding the linker to the cysteine 34 thiol of the albumin. In some embodiments, the exendin-4 albumin conjugate is at a concentration of about 10 mg/ml to 20 mg/ml. In some embodiments, the buffer is a succinate, maleate, citrate-phosphate, histidine (e.g., L-histidine), a phosphate (e.g., sodium phosphate), or acetate (e.g., sodium acetate) buffer or combinations thereof with a pH of about 4.0 to 8.0. In some embodiments, the tonicity modifier is sodium chloride. In some embodiments, the stabilizer is sodium octanoate or myristic acid. In some embodiments, the surfactant is pluronic F68.
[0322] In certain embodiments, the pharmaceutical formulation comprises, or alternatively consists of, about 1 mg/ml to about 15 mg/ml insulinotropic peptide conjugate in 5-30 mM sodium phosphate buffer at about pH 5.0-8.0 containing 100-200 mM sodium chloride, 1-10 mM sodium octanoate, and 1-30 mg/L polysorbate 80. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml insulinotropic peptide conjugate in 5-30 mM sodium phosphate buffer at about pH 5.0-8.0 containing 100-200 mM sodium chloride, 1-10 mM sodium octanoate, and 1-30 mg/L polysorbate 80. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml insulinotropic peptide conjugate in 10 mM sodium phosphate buffer containing 100-200 mM sodium chloride, 1-10 mM sodium octanoate, and 1-30 mg/L polysorbate 80 wherein said formulation has a pH of about 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 100-200 mM sodium chloride, 1-10 mM sodium octanoate, and 1-30 mg/L polysorbate 80. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 135 mM sodium chloride, 1.6 mM sodium octanoate, and 15 mg/L polysorbate 80.
[0323] In some embodiments, the pharmaceutical formulation comprises, or alternatively consists of, about 1 mg/ml to about 15 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM sodium phosphate buffer at about pH 5.0-8.0 containing 100-200 mM sodium chloride, 1-10 mM sodium octanoate, and 1-30 mg/L polysorbate 80. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM sodium phosphate buffer at about pH 5.0-8.0 containing 100-200 mM sodium chloride, 1-10 mM sodium octanoate, and 1-30 mg/L polysorbate 80. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml exendin-4(l-39) Lys40 (E-AEEA-MPA)-NH2 albumin conjugate in 10 mM sodium phosphate buffer containing 100-200 mM sodium chloride, 1-10 mM sodium octanoate, and 1-30 mg/L polysorbate 80 wherein said formulation has a pH of about 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 100-200 mM sodium chloride, 1-10 mM sodium octanoate, and 1-30 mg/L polysorbate 80. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 135 mM sodium chloride, 1.6 mM sodium octanoate, and 15 mg/L polysorbate 80.
[0324] In a particular embodiment, the formulation consists of about 1 mg/ml to about 15 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 135 mM sodium chloride, 1.6 mM sodium octanoate, and 15 mg/L polysorbate 80. In a particular embodiment, the formulation consists of about 1 mg/ml to about 15 mg/ml of a conjugate of albumin to exendin-4, or a derivative therof, in 10 mM sodium phosphate buffer at about pH 7.0 containing 135 mM sodium chloride, 1.6 mM sodium octanoate, and 15 mg/L polysorbate 80. In a particular embodiment, the formulation consists of about 1 mg/ml to about 15 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 135 mM sodium chloride, 1.6 mM sodium octanoate, and 15 mg/L polysorbate 80. In a particular embodiment, the formulation consists of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 135 mM sodium chloride, 1.6 mM sodium octanoate, and 15 mg/L polysorbate 80.
[0325] In certain embodiments, the pharmaceutical formulation comprises, or alternatively consists of, about 1 mg/ml to about 15 mg/ml insulinotropic peptide conjugate in 5-30 mM sodium acetate buffer at about pH 4.5-5.5, containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, and either 100-200 mM sodium chloride or 2-8% (w/v) sorbitol. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml insulinotropic peptide conjugate in 5-30 mM sodium acetate buffer at about pH 4.5-5.5, containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, either 100-200 mM sodium chloride or 2-8% (w/v) sorbitol, and optionally a preservative. In a particular
embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml insulinotropic peptide conjugate in 10 mM sodium acetate buffer containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, either 100-200 mM sodium chloride or 2-8% (w/v) sorbitol, and optionally a preservative, wherein said formulation has a pH of about 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, or 5.5. In a particular embodiment, the formulation comprises or alternatively consists of, 10 mg/ml insulinotropic peptide conjugate in 10 mM sodium acetate buffer at about pH 5.0 containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, and either 100- 200 mM sodium chloride or 2-8% (w/v) sorbitol. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml insulinotropic peptide conjugate in 10 mM sodium acetate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68 (i.e., poloxamer 188).
[0326] In certain embodiments, the pharmaceutical formulation comprises, or alternatively consists of, about 1 mg/ml to about 15 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM sodium acetate buffer at about pH 4.5-5.5, containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, either 100-200 mM sodium chloride or 2-8% (w/v) sorbitol, and optionally a preservative. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM sodium acetate buffer at about pH 4.5-5.5, containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, either 100-200 mM sodium chloride or 2-8% (w/v) sorbitol, and optionally a preservative. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml exendin-4(l-39) Lys (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium acetate buffer containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, and either 100-200 mM sodium chloride or 2-8% (w/v) sorbitol wherein said formulation has a pH of about 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, or 5.5. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium acetate buffer at about pH 5.0 containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, and either 100-200 mM sodium chloride or 2-8% (w/v) sorbitol. In a particular embodiment, the formulation comprises, or alternatively consists of, 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM sodium acetate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68 (i.e., poloxamer 188), and optionally a preservative.
[0327] In a particular embodiment, the formulation consists of about 1 mg/ml to about 15 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium acetate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68 (i.e., poloxamer 188). In a particular embodiment, the formulation consists of about 1 mg/ml to about 15 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM sodium acetate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68 (i.e., poloxamer 188). In a particular embodiment, the formulation consists of about 1 mg/ml to about 15 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium acetate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68 (i.e., poloxamer 188). In a particular embodiment, the formulation consists of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM sodium acetate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68 (i.e., poloxamer 188), and optionally a preservative.
[0328] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml insulinotropic peptide conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, and optionally a preservative, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 4.5. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 6.5.
[0329] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, and optionally a preservative, wherein the formulation has a pH of about4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 4.5. In a particular embodiment, the formulation comprises 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation comprises 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 6.5.
[0330] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml insulinotropic peptide conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, and optionally a preservative, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 4.5. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation comprises 10 mg/ml insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 6.5. [0331] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, and optionally a preservative, wherein the formulation has a pH of about4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 4.5. In a particular embodiment, the formulation comprises 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation comprises 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 6.5.
[0332] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 4.5 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 15 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, 160 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 15 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 4.5. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of 5.5. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 6.5.
[0333] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 4.5. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 160 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 6.5.
[0334] In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 6.5. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- ΜΡΑ)-Ν¾ albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε- AEEA-MPA)-NH2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 mM, wherein the formulation has a pH of about 6.5.
[0335] In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 6.5. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε- AEEA-MPA)-NH2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, optionally a preservative, and optionally a buffer, wherein when present, the buffer is at a concentration of no more than 0.05 M, wherein the formulation has a pH of about 6.5.
[0336] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate and 0.1% (w/v) pluronic F68, wherein the formulation has a pH of about 6.5.
[0337] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate, 150 mM sodium chloride, 15 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, 150 mM sodium chloride, 15 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 4.0 to 7.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 5.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 5.5. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 6.0. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate, 150 mM sodium chloride, 15 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative, wherein the formulation has a pH of about 6.5.
[0338] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 5-30 mM succinate buffer at about pH 4.0-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 5-30 mM succinate buffer at about pH 4.0-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM succinate buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM succinate buffer at about pH 4.0-7.0 containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM succinate buffer at about pH 4.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM succinate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM succinate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM succinate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM succinate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative.
[0339] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM succinate buffer at about pH 4.0 to 7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM succinate buffer at about pH 4.0 to 7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε- AEEA-MPA)-NH2 albumin conjugate in 10 mM succinate buffer at about pH 4.0 to 7.0 containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 4.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative.
[0340] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM succinate buffer at about pH 4.0 to 7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of exendin-4, or a derivative thereof, in 10 mM succinate buffer at about pH 4.0 to 7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM succinate buffer at about pH 4.0 to 7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 4.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68.
[0341] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM succinate buffer at about pH 4.0 to 7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of exendin-4, or a derivative thereof, in 10 mM succinate buffer at about pH 4.0 to 7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) plutonic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 4.0 to 7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 4.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM succinate buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM succinate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative.
[0342] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 5-30 mM maleate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 5-30 mM maleate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM maleate buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, and 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM maleate buffer at about pH 5.5-7.0 containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM maleate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM maleate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM maleate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative.
[0343] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM maleate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM maleate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 5.5-7.0 containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. [0344] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM maleate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM maleate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε- AEEA-MPA)-NH2 albumin conjugate in 10 mM maleate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68.
[0345] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM maleate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM maleate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM maleate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM maleate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative.
[0346] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 5-30 mM citrate-phosphate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100- 200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 5-30 mM citrate- phosphate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM citrate-phosphate buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM citrate-phosphate buffer at about pH 5.5-7.0 containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM citrate-phosphate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM citrate-phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM citrate-phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative.
[0347] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM citrate-phosphate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM citrate -phosphate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate-phosphate buffer containing 1- 20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 5.5-7.0 containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate -phosphate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative.
[0348] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM citrate-phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate -phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM citrate- phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68.
[0349] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM citrate-phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)- NH2 albumin conjugate in 10 mM citrate -phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM citrate-phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM citrate-phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative.
[0350] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 5-30 mM L-histidine buffer at about pH 5.0-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 5-30 mM L-histidine buffer at about pH 5.0-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100- 200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM L-histidine buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 5.0, 5.1, 5.2, 5.3, 5.4 , 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM L-histidine buffer at about pH 5.0-7.0 containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM L-histidine buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM L-histidine buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM L-histidine buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM L-histidine buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a
preservative.
[0351] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM L- histidine buffer at about pH 5.0-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM L-histidine buffer at about pH 5.0-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 5.0, 5.1, 5.2, 5.3, 5.4 , 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 5.0-7.0 containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM L-histidine buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative.
[0352] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM L-histidine buffer at about pH 5.0-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM L-histidine buffer at about pH 5.0-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε- AEEA-MPA)-NH2 albumin conjugate in 10 mM L-histidine buffer at about pH 5.0-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L- histidine buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68.
[0353] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM L-histidine buffer at about pH 5.0-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM L-histidine buffer at about pH 5.0-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L- histidine buffer at about pH 5.0-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM L-histidine buffer at about pH 5.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 5.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM L-histidine buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative.
[0354] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 5-30 mM sodium phosphate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100- 200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 5-30 mM sodium phosphate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and optionally a preservative.
[0355] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM sodium phosphate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 5-30 mM sodium phosphate buffer at about pH 5.5-7.0, containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer containing 1-20 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100- 200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 1-15 mM sodium octanoate, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate 0.1% (w/v) pluronic F68, and optionally a preservative.
[0356] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate and 0.1% (w/v) pluronic F68.
[0357] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 5.5-7.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 5 mM sodium octanoate, 0.1% (w/v) pluronic F68, and a preservative.
[0358] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 5-30 mM sodium phosphate buffer at about pH 6.0-8.0, containing 0.01-5.0 mM myristic acid, 0.05 to 0.2% (w/v) pluronic F68, 100- 200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 5-30 mM sodium phosphate buffer at about pH 6.0-8.0, containing 0.01-5.0 mM myristic acid, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer containing 0.01-5.0 mM myristic acid, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 6.5-8.0 containing 0.01-5.0 mM myristic acid, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml of an
insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 7.5 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and optionally a preservative.
[0359] In some embodiments, the pharmaceutical formulation comprises about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM sodium phosphate buffer at about pH 6.0-8.0, containing 0.01-5.0 mM myristic acid, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 5-30 mM sodium phosphate buffer at about pH 6.0-8.0, containing 0.01-5.0 mM myristic acid, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer containing 0.01-5.0 mM myristic acid, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative, wherein said formulation has a pH of about 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.5-8.0 containing 0.01-5.0 mM myristic acid, 0.05 to 0.2% (w/v) pluronic F68, 100-200 mM sodium chloride, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and optionally a preservative. In a particular embodiment, the formulation comprises 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.5 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and optionally a preservative.
[0360] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 6.0-8.0 containing 150 mM sodium chloride, 0.1 mM myristic acid and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM sodium phosphate buffer at about pH 6.0-8.0 containing 150 mM sodium chloride, 0.1 mM myristic acid and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l- 39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.0-8.0 containing 150 mM sodium chloride, 0.1 mM myristic acid and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consist of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 0.1 mM myristic acid and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consist of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 0.1 mM myristic acid and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consist of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 150 mM sodium chloride, 0.1 mM myristic acid and 0.1% (w/v) pluronic F68. In a particular embodiment, the formulation consist of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.5 containing 150 mM sodium chloride, 0.1 mM myristic acid and 0.1% (w/v) pluronic F68.
I l l [0361] In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of an insulinotropic peptide conjugate in 10 mM sodium phosphate buffer at about pH 6.0-8.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml of a conjugate of albumin to exendin-4, or a derivative thereof, in 10 mM sodium phosphate buffer at about pH 6.0-8.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consists of or consists essentially of about 1 mg/ml to about 100 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.0-8.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consist of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ- MPA)-NH2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consist of or consists essentially of 10 mg/ml exendin- 4(1-39) Lys40 (ε-ΑΕΕΑΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 6.5 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consist of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.0 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and a preservative. In a particular embodiment, the formulation consist of or consists essentially of 10 mg/ml exendin-4(l-39) Lys40 (8-AEEA- MPA)-NH2 albumin conjugate in 10 mM sodium phosphate buffer at about pH 7.5 containing 150 mM sodium chloride, 0.1 mM myristic acid, 0.1% (w/v) pluronic F68, and a preservative.
[0362] The pharmaceutical formulations provided herein can be in any form deemed useful to those of skill in the art. For instance, they can be in the form of liquid or lyophilized formulations, unit dosage forms or multi-use dosage forms and combinations thereof. Thus, the formulations include liquid unit dosage forms, liquid multi-use forms, lyophilized unit dosage forms and lyophilized multi-use dosage forms.
[0363] In some embodiments, the formulation is a liquid formulation. In other embodiments, the formulation is a lyophilized formulation. Lyophilization is a commonly employed technique for preserving proteins which serves to remove water from the peptide preparation of interest. An excipient can be included in pre -lyophilized formulations to enhance stability during the freeze- drying process and/or to improve stability of the lyophilized product upon storage. See Pikal, M. 1990, Biopharm. 3(9):26-30 and Arakawa et al. 1991, Pharm. Res. 8(3):285-291. [0364] Lyophilized formulations can be reconstituted according to the judgment of those of skill in the art. In preferred embodiments, a lyophilized formulation is provided which, when reconstituted, e.g., with water for injection, results in one of the liquid formulations described herein. The present invention also provides a method of reconstituting a lyophilized formulation of an insulinotropic peptide conjugate comprising providing the lyophilized formulation, and reconstituting the lyophilized formulation to form an insulinotropic peptide conjugate formulation described herein. In certain embodiments, the lyophilized formulation is provided in a 2-chamber syringe and reconstituted in the syringe. In particular embodiments, the lyophilized formulation is provided in a 2-chamber cartridge and reconstituted in the cartridge.
[0365] At the desired stage, typically when it is time to administer the peptide to the subject, the lyophilized formulation can be reconstituted with a diluent such that the protein concentration in the reconstituted formulation is at least 1, 2, 3, 4, 5, 10, 20, 30, 40, 50 mg/ml. In some embodiments, the protein concentration in the reconstituted formulation is from about 1 mg/ml to about 100 mg/ml, from about 1 mg/ml to about 50 mg/ml, or from about 1 mg/ml to about 15 mg/ml. In particular embodiments, the lyophilized formulation can be reconstituted with a diluent such that the protein concentration in the reconstituted formulation is about 45-55 mg/ml. In preferred embodiments, the lyophilized formulation can be reconstituted with a diluent such that the protein concentration in the reconstituted formulation is about 50 mg/ml. The diluent can be any diluent deemed suitable by one of skill, e.g., water for injection, and the like.
[0366] The pharmaceutical formulations provided herein include both unit dosage forms and multi-use dosage forms. In some embodiments, the formulations are in unit dosage forms. " Unit dosage form" refers to a packaged form of the pharmaceutical formulation in an amount that is intended for a single administration to a subject. In some embodiments, the formulations are in unit dosage forms. In certain embodiments, the unit dosage comprises about 10-100,000 μg, 100- 50,000 μg, 1000-10,000 μg, or 1000-5000 μg insulinotropic peptide conjugate. In particular embodiments, the unit dosages comprise about 100, 200, 300, 400, 500, 750, 1000, 20,000, 30,000, 40,000, 50,000, 75,000 or 100,000 μg insulinotropic peptide conjugate. Such unit dosages can be prepared according to techniques known to those of skill in the art.
[0367] In some embodiments, the formulations are in multi-use dosage forms. Multi-use formulations can facilitate ease of use for subjects, reduce waste by allowing complete use of vial contents and result in significant cost savings for manufacture. Multi-use pharmaceutical formulations can be contained in multi-dose containers, e.g., vials, ampoules, etc., that allow for the extraction of partial amounts of the formulations at various times. One or more preservatives compatible with the buffer in the formulations can be present in multi-use formulations as described in detail above. [0368] Preferably, the formulations of the present invention are stable. In some embodiments, the formulations are stable for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36 or more than 36 months at a temperature of about 4 °C. In other embodiments, the formulations are stable for at least about 1, 2 or 3 weeks, or at least about 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,
21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, or more than 36 months at a temperature of about 25 °C. In other embodiments, the formulations are stable for at least about 1, 2 or 3 weeks, or at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21,
22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, or more than 36 months at a temperature of about 40 °C.
VII. Kits
[0369] In a further embodiment, the present invention provides kits comprising an
insulinotropic peptide or modified insulinotropic peptide, which can be used, for instance, in practicing the methods of treatment described herein. For example, the present invention provides kits for the treatment of type II diabetes mellitus in a subject in need thereof. The kits comprise an insulinotropic peptide or modified insulinotropic peptide in a package for distribution to a practitioner of skill in the art. The kits can comprise a label or labeling with instructions for use of the insulinotropic conjugate as described herein, e.g, instructions for administering the insulinotropic peptide or modified insulinotropic peptide for the treatment of subjects with (or who are or are undergoing), e.g. pre-diabetes (e.g., impaired glucose tolerance (IGT) and impaired fasting glucose (IFG)), diabetes, e.g., type I diabetes or type II diabetes, late
autoimmune diabetes in adults ("LAD A" ) also known as late onset autoimmune diabetes of adulthood, slow onset type I diabetes and type 1.5 diabetes, steroid induced diabetes, Human Immunodeficiency Virus (HIV) Treatment-Induced Diabetes, diabetes development in subjects with congenital or HIV-Associated Lipodystrophy ("Fat Redistribution Syndrome"), obesity (i.e., BMI of 30 kg/m2 or greater), overweight (i.e., BMI between 25 kg/m2 and 30 kg/m2), metabolic syndrome (Syndrome X), nervous system disorders, insulin resistance, hypoglycemia
unawareness, restrictive lung disease, a gastrointestinal disorder, e.g., irritable bowel syndrome (IBS), functional dyspepsia, pain associated with gastrointestinal disorders, e.g., pain associated with IBS and functional dyspepsia, inflammatory bowel disease (IBD), e.g., Crohn's disease and ulcerative colitis, pain associated with IBD, hyperglycemia, e.g., hyperglycemia associated with surgery (e.g., a major surgical procedure, e.g., coronary bypass surgery) e.g., hyperglycemia associated with surgery on subjects with diabetes, e.g., type II diabetes, metabolic syndrome, coronary heart failure (CHF), disorders associated with beta cell disfunction, disorders associated with the absence of beta cells, disorders associated with insufficient numbers of beta cells, and other conditions treatable with an insulinotropic peptide or insulinotropic peptide conjugate.
[0370] The kits can comprise a label or labeling with instructions for use of the insulinotropic peptide or modified insulinotropic peptide, e.g, instructions for administering the insulinotropic peptide or modified insulinotropic peptide in accordance with the administration regimens provided herein, to promote weight loss, stimulate insulin synthesis and release, to enhance adipose, muscle or liver tissue sensitivity toward insulin uptake, to stimulate glucose uptake, to slow (e.g., decrease the rate of) digestive processes, e.g., gastric emptying, to block or inhibit secretion of glucagon, to promote beta cell function, proliferation, and/or activity, to restore first phase insulin release in subjects with diabetes, to reduce food intake, to reduce appetite, to prevent or protect against liver disease, e.g., liver disease associated with obesity, diabetes, or hyperglycemia (e.g., non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH)).
[0371] The instructions on the label can further include instructions for storage conditions of the insulinotropic peptide or modified insulinotropic peptide.
[0372] In certain embodiments, the kit can comprise one or more containers, e.g., bottles, vials, ampoules, cartridges, pre-filled containers, e.g., pre -filled syringes or prefilled injection pens, microchip (e.g., a microchip for controlled release of its contents) or test tubes which contain a unit dosage or a multi-use dosage of the insulinotropic peptide or modified insulinotropic peptide formulation. In particular embodiments, components of the insulinotropic peptide or modified insulinotropic peptide formulation are contained in more than one, i.e., in different containers. In some embodiments, the dosage forms can be contained as liquid or lyophilized formulations. Kits comprising lyophilized dosage forms can further comprise one or more additional containers comprising a diluent for reconstituting the lyophilized formulation, such that the protein, i.e., insulinotropic peptide or modified insulinotropic peptide, concentration in the reconstituted formulation is at least 1, 2, 3, 4, 5, 10, 20, 30, 40, 50 mg/ml, for example from about 1 mg/ml to about 100 mg/ml, more preferably from about 1 mg/ml to about 50 mg/ml, and most preferably from about 1 mg/ml to about 15 mg/ml. In certain embodiments, the diluent for reconstituting the lyophilized formulation is provided in a 2-chamber syringe also containing the lyophilized formulation, and reconstitution is performed in the syringe. In certain embodiments, the diluent for reconstituting the lyophilized formulation is provided in a 2-chamber cartridge also containing the lyophilized formulation, and reconstitution is performed in the cartridge.
[0373] The kit can further comprise one or more additional components useful for carrying out the methods described herein, including, but not limited to, buffers, filters, needles, syringes, and package inserts with instructions for use. In a particular embodiment, the kit comprises a needle, e.g., a 23-gauge needle, a 24-gauge needle, a 25-gauge needle, a 26-gauge needle, a 27-gauge needle, a 28-gauge needle, a 29-gauge needle, a 30-gauge needle, a 31 -gauge needle, a 32-gauge needle, or a 33 -gauge needle, or a higher gauge needle, useful, e.g. for the subcutaneous administration of the insulinotropic peptide or modified insulinotropic peptide formulation to a subject. In certain embodiments, the kits can comprise components useful for the safe disposal of means for administering the insulinotropic peptide or modified insulinotropic peptide formulation, e.g. a sharps container for used syringes and needles.
[0374] In a preferred embodiment, the kit comprises one or more syringes pre-loaded with a first dosage of the insulinotropic peptide or modified insulinotropic peptide formulation, and one or more syringes pre-loaded with a second higher dosage, of the insulinotropic peptide or modified insulinotropic peptide, useful e.g., for administering increasing dosages to a subject during the course of a administration regimen described herein. In a particular embodiment, the kit comprises 1, 2, 3, 4, 5, 6, 7, 8, or more than 8 syringes pre-loaded with a first dosage of the insulinotropic peptide or modified insulinotropic peptide formulation. In another particular embodiment, the kit comprises 1, 2, 3, 4, 5, 6, 7, 8, or more than 8 syringes pre-loaded with a second higher dosage of the insulinotropic peptide or modified insulinotropic peptide formulation.
[0375] In other embodiments, the kit comprises one, two, three, four, five, six, seven, eight, nine, ten or more than ten empty syringes, and one, two, three, four, five, six, seven, eight, nine, ten or more than ten vials, wherein each vial contains 1 dose, 2 doses, 3 doses, 4 doses, 5 doses, 6 doses, 7 doses, 8 doses, 9 doses, 10 doses or more than 10 doses of the insulinotropic peptide or modified insulinotropic peptide formulation. In other embodiments, the kit comprises one, two, three, four, five, six, seven, eight, nine, ten or more than ten syringes pre-loaded with 1 dose, 2 doses, 3 doses, 4 doses, 5 doses, 6 doses, 7 doses, 8 doses, 9 doses, 10 doses, or more than 10 doses of the insulinotropic peptide or modified insulinotropic peptide formulation. In some embodiments, the syringe comprises a luer-lock, luer-cone, or other needle fitting connector that facilitates attachment of a disposable needle. In other embodiments, the syringe comprises a staked, i.e., permanent, needle.
[0376] In a particular embodiment, the kit comprises a pen-type delivery apparatus and one, two, three, four, five, six, seven, eight, nine, ten or more than ten replaceable cartridges, wherein the replaceable cartridge comprises, e.g., is pre-loaded with 1 dose, 2 doses, 3 doses, 4 doses, 5 doses, 6 doses, 7 doses, 8 doses, 9 doses, 10 doses or more than 10 doses of the insulinotropic peptide or modified insulinotropic peptide formulation. In some embodiments, the formulation is provided as a liquid formulation in a single chamber syringe or cartridge. In some embodiments, the formulation is provided as a lyophilized formulation in a two-chamber syringe or cartridge, and reconstitution is performed in the syringe or cartridge. In certain embodiments where the pen- type delivery apparatus comprises multiple doses, the dose can be pre-set, i.e., fixed. In other embodiments, the dose can be a flexible dose, i.e., dialed-in by the user. In a particular embodiment, the kit comprises one, two, three, four, five, six, seven, eight, nine, ten or more than ten pen-type delivery apparatuses pre-loaded with one, two, three, four, five, six, seven, eight, nine, ten or more than ten doses of the insulinotropic peptide or modified insulinotropic peptide formulation. In some embodiments, the pen-type delivery apparatus comprises a luer-lock, luer- cone, or other needle fitting connector that facilitates attachment of a disposable needle. In a particular embodiment, the kit comprises a disposable pen-type delivery apparatus. In other embodiments, the pen-type delivery apparatus comprises a staked, i.e., permanent, needle.
VIII. Exemplar Embodiments
1. A method of treating a human subject having a fasting blood glucose level characteristic of the presence of diabetes, said method comprising:
(a) determining a value of the fasting blood glucose level of said subject, by a method comprising measuring said fasting blood glucose level, wherein said determining is performed by said subject;
(b) within 24 hours of said determining step, administering to the subject an amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity; wherein said administering is performed by said subject using a flexible dosage device, and wherein said administering is the first administering of said compound that occurs subsequent to said determining step;
(c) after said administering step, repeating steps (a) and (b) for a plurality of cycles, wherein each occurrence of said administering step in a cycle is with said amount being increased relative to the amount of said compound administered in the most recent cycle of said
determining and administering steps, unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject; wherein if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject. 2. The method of embodiment 1 , wherein said fasting blood glucose level characteristic of the presence of diabetes is a level of greater than 125 mg/dL.
3. A method of treating a human subject having a fasting blood glucose level characteristic of the presence of pre-diabetes, said method comprising:
(a) determining a value of the fasting blood glucose level of said subject, by a method comprising measuring said fasting blood glucose level, wherein said determining is performed by said subject;
(b) within 24 hours of said determining step, administering to the subject an amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity; wherein said administering is performed by said subject using a flexible dosage device, and wherein said administering is the first administering of said compound that occurs subsequent to said determining step;
(c) after said administering step, repeating steps (a) and (b) for a plurality of cycles, wherein each occurrence of said administering step in a cycle is with said amount being increased relative to the amount of said compound administered in the most recent cycle of said determining and administering steps, unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of pre-diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject; wherein if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
4. The method of embodiment 3, wherein said fasting blood glucose level characteristic of the presence of pre-diabetes is a level in the range of 100 mg/dL to 125 mg/dL.
5. The method of any one of embodiments 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 80 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
6. The method of any one of embodiments 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 75 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
7. The method of any one of embodiments 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 70 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
8. The method of any one of embodiments 1 to 4, wherein if during step (c), the subject experiences a condition of diarrhea or dyspepsia intolerable to the subject, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
9. The method of embodiment 1 or 2, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 70 to 125 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
10. The method of embodiment 1 or 2, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 80 to 125 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
11. The method of any one of embodiments 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 70 to 99 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
12. The method of any one of embodiments 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 80 to 99 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
13. The method of any one of embodiments 1 to 12, wherein said value of the fasting blood glucose level of the subject is an average of a plurality of measures of the fasting blood glucose level of said subject over a period of time, wherein said plurality of measures comprises said measure obtained in said measuring step. 14. The method of any one of embodiments 1 to 12, wherein said value of the fasting blood glucose level of the subject is the measure of the fasting blood glucose level of said subject obtained in said measuring step.
15. The method of any one of embodiments 1 to 14, wherein said administering of step (b) is performed within three hours of determining the fasting blood glucose level of the subject in step (a).
16. The method of any one of embodiments 1 to 15, wherein each cycle of (c) comprises performing said determining and administering steps once every 1, 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, or 13 days, or once a week, or once every 2, 3, or 4 weeks, or once a month.
17. The method of any one of embodiments 1 to 16, wherein each cycle of step (c) comprises administering said dose within three hours of determining the fasting blood glucose level of the subject.
18. The method of any one of embodiments 1 to 17, wherein said repeating of step (c) comprises decreasing said dose relative to the most recently administered dose.
19. The method of any one of embodiments 1 to 18, wherein said repeating of step (c) comprises repeating said determining and administering steps for at least 10 cycles.
20. The method of any one of embodiments 1 to 19, wherein the modified insulinotropic peptide has an increased half-life relative to the non-modified insulinotropic peptide.
21. The method of any one of embodiments 1 to 20, wherein the modified insulinotropic peptide is an insulinotropic peptide conjugate.
22. The method of any one of embodiments 1 to 21, wherein the insulinotropic peptide is selected from the group consisting of a glucagon-like-peptide 1 (GLP-1) peptide, an exendin peptide, and analogs thereof.
23. The method of embodiment 22, wherein the insulinotropic peptide is exendin-4 or an analog thereof.
24. The method of any one of embodiments 1 to 23, wherein the modified insulinotropic peptide is a conjugate of albumin and an exendin peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence.
25. The method of any one of embodiments 1 to 24, wherein said administering the modified insulinotropic peptide comprises administering a pharmaceutical formulation comprising: a conjugate of albumin and an exendin peptide, said exendin peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence, said conjugate being at a concentration of about 1 mg/ml to about 100 mg/ml; optionally a buffer; a tonicity modifier, wherein the tonicity modifier is at a concentration of at least 1 mM; a stabilizer; a surfactant, and optionally a preservative, wherein said formulation has a pH from about 4 to about 8.
26. The method of embodiment 25, wherein the buffer is at a concentration of no more than 0.05 mM, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
27. The method of embodiment 25, wherein the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
28. The method of embodiment 25, wherein the buffer is at a concentration of no more than 0.05 mM, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5.
29. The method of embodiment 25, wherein the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5.
30. The method of embodiment 25, wherein the buffer is at a concentration of no more than 0.05 mM, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
31. The method of embodiment 25, wherein the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
32. The method of embodiment 25, wherein the buffer is at a concentration of no more than 0.05 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 15 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
33. The method of embodiment 25, wherein the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 15 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0. 34. The method of embodiment 25, wherein the buffer is L-histidine at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5.
35. The method of embodiment 25, wherein the buffer is L-histidine at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
36. The method of embodiment 25, wherein the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
37. The method of embodiment 25, wherein the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5.
38. The method of embodiment 25, wherein the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
39. The method of embodiment 25, wherein the buffer is sodium acetate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
40. The method of any one of embodiments 24 to 39, wherein the conjugate comprises albumin cysteine 34 thiol covalently linked to a [2-[2-[2- maleimidopropionamido(ethoxy)ethoxy] acetic acid linker covalently linked to the epsilon amino of a lysine of said exendin peptide.
41. The method of embodiment 40, wherein said lysine has been added to the native exendin- 4 sequence.
42. The method of embodiment 40, wherein said lysine has been added to the carboxy terminus of the native exendin-4 sequence.
43. The method of embodiment 40, wherein the conjugate is according to the following:
Figure imgf000124_0001
(SEQ ID NO: 31)
wherein X is S, O, or NH of an amino acid of albumin.
44. The method of embodiment 43, wherein X is cysteine 34 thiol of albumin.
45. The method of any one of embodiments 24 to 44, wherein the albumin is human serum albumin.
46. The method of any one of embodiments 24 to 44, wherein the albumin is recombinant human albumin.
47. The method of any one of embodiments 24 to 46, wherein the conjugate comprises recombinant human serum albumin cysteine 34 thiol covalently linked to a [2- [2- [2
maleimidopropionamido(ethoxy)ethoxy] acetic acid linker covalently linked to the epsilon amino of the carboxy terminal lysine of exendin-4(l-39)Lys40-NH2.
48. The method of any one of embodiments 24 to 47, wherein said conjugate is at a concentration from about 1 mg/ml to about 50 mg/ml.
49. The method of any one of embodiments 24 to 47, wherein said conjugate is at a concentration from about 1 mg/ml to about 15 mg/ml.
50. The method of any one of embodiments 24 to 47, wherein said conjugate is at a concentration from about 1 mg/ml to about 10 mg/ml.
51. The method of any one of embodiments 24 to 47, wherein said conjugate is at a concentration of about 10 mg/ml.
52. The method of any one of embodiments 24 to 47, wherein said conjugate is at a concentration of about 20 mg/ml.
53. The method of any one of embodiments 24 to 52, wherein said dose of step (b) is at least about 0.5 mg of the conjugate.
54. The method of any one of embodiments 24 to 52, wherein said dose of step (b) is about 1.5 mg of the conjugate.
55. The method of any one of embodiments 24 to 54, wherein each cycle of step (c) comprises administering a dose of the conjugate which is about 0.1, 0.2, 0.3, 0.4 or 0.5 mg greater than the most recent prior administered dose, provided, however, that said dose does not exceed about 5.0 mg of the conjugate. 56. The method of any one of embodiments 1 to 55, wherein said administering comprises subcutaneously administering the pharmaceutical formulation to the subject.
57. The method of any one of embodiments 1 to 56, wherein said administering comprises administering the pharmaceutical formulation to the subject with a pen-type delivery apparatus.
58. The method of embodiment 57, wherein said pen-type delivery apparatus comprises multiple doses of the pharmaceutical formulation.
59. The method of any one of embodiments 1 to 58, wherein the subject is a human.
60. The method of any one of embodiments 1 to 59, wherein the subject has been on a stable dose of metformin of > 1000 mg metformin daily for at least 3 months.
EXAMPLES
[0377] The invention is illustrated by the following examples which are not intended to be limiting in any way.
Example 1: Preparation of Exendin-4 Albumin Conjugates
[0378] Exendin-4( 1-39) Lys40 (ε- AEEA-MPA)-NH2 conjugated with recombinant human albumin Cys34 (hereinafter " exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin-conjugate" in the following examples) was prepared as described in detail in U.S. Pat. No. 6,329,336; U.S. Pat. Pub. No. 2005/0267293; U.S. Pat. App. No. 11/645,297, filed December 22, 2006, entitled "Process for the Production of Preformed Conjugate of Recombinant Albumin," the contents of each of which are incorporated by reference herein in their entireties.
Preparation of Exendin-4(l-39) Lys40 ( ε-ΑΕΕΑ-ΜΡΑ )-NH2
[0379] Exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 was prepared according to methods described in U.S. Pat. No. 6,329,336, which is incorporated by reference herein in its entirety. Briefly, solid phase peptide synthesis of Exendin-4 on a 100 μιηοΐε scale was performed using manual solid-phase synthesis and a Symphony Peptide Synthesizer using Fmoc protected Rink Amide MBHA resin. The selective deprotection of the Lys(Aloc) group was performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHC13 NMM:HOAc (18:1:0.5) for 2 h. The resin was then washed with CHC13 (6X 5 mL), 20% HOAc in DCM (6X5 mL), DCM (6X5 mL), and DMF (6X5 mL). The synthesis was then re-automated for the addition of the aminoethoxyethoxyacetic acid (AEEA) group the 3- maleimidopropionic acid (MP A). Resin cleavage and product isolation was performed using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry- ice cold Et2 O. The product was purified by preparative reverse phase HPLC using a Varian (Rainin) preparative binary HPLC system.
Preparation of Exendin-4( 1 -39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin-Conjugates [0380] Exendin-4( 1 -39) Lys4U (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 was then conjugated to human
recombinant serum albumin as described in U.S. Pat. App. No. 11/645,297 (Publication No. 2007/0269863), filed December 22, 2006, entitled "Process for the Production of Preformed Conjugates of Albumin and a Therapeutic Agent," the contents of which are incorporated by reference herein in their entirety. Recombinant albumin expressed in Saccharomyces cerevisiae was purified and treated with thioglycolic acid, and purified by phenyl sepharose HIC prior to conjugation. The conjugation reaction comprised 35 μΐ of 10 mM exendin-4(l-39) Lys40 (ε- AEEA-MPA)-NH_2 combined with 175 μΐ of mercaptalbumin enriched albumin in at a final molar ratio of 0.7: 1. The reaction proceeded for 30 minutes at 37 °C, and was then stored at 4 C for liquid chromatography / mass spec analysis and purification by butyl sepharose HIC.
[0381] Exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin-conjugate was purified by loading the conjugation reaction mixture onto a hydrophobic support equilibrated in aqueous buffer having a high salt content; applying to the support a gradient of decreasing salt concentration; and collecting the eluted albumin conjugate as described in U.S. Pat. App. No. 11/645,297
(Publication No. 2007/0269863), filed December 22, 2006, entitled "Process for the Production of Preformed Conjugates of Albumin and a Therapeutic Agent," which is incorporated by reference herein in its entirety.
Example 2: Treatment of Type II Diabetes, Based On A "Titrate-To-Target" Treatment Schedule, With an Exendin-4(l-39) Lys40 (s-AEEA-MPA)-NH2 Albumin-Coniugate
Formulation
[0382] This example describes a 12-week randomized, placebo-controlled, double -blind escalating dose Phase lib clinical study based on a titrate-to-target treatment schedule to evaluate the maximum tolerable and effective dose of an exendin-4(l-39) Lys40 (ε-ΑΕΕΑΜΡΑ)-ΝΗ2 albumin-conjugate formulation administered subcutaneously to subjects with Type II diabetes mellitus. The conjugate is prepared as described in Example 1 and is administered at a concentration of 10 mg/ml in a formulation described herein. The starting dose of the conjugate is 1.5 mg, and is titrated weekly at 0.5 mg increments to a maximum weekly dose of 5.0 mg (0.5 ml), as follows:
Figure imgf000126_0001
6 4.0 mg 0.40 ml
7 4.5 mg 0.45 ml
8 5.0 mg 0.50 ml
[0383] For each subject, the maximum dose administered from weeks 1-8 is then administered for the remainder of the study. Subjects receiving placebo are dosed according to the same schedule, i.e., with an increasing weekly injection volume, as described above.
[0384] Fasting plasma glucose levels are determined prior to administering each dose
(including prior to administering the first dose), and on each of days 2-6 following each dose of the conjugate. For each subject, the scheduled weekly increase in dosage of the conjugate is subject to the following conditions (based on the daily fasting glucometer readings and the patient's subjective experience from the previous week):
(1) attaining a glucose target of between 100-125 mg/dL (based on mean weekly fasting glucose);
(2) attaining a post-dosing fasting glucose level of < 70 mg/dL following the dose of the previous week; and
(3) experiencing any vomiting or nausea intolerable to the subject following the dose of the previous week, or experiencing any vomiting or moderate nausea for two consecutive doses.
[0385] In the event that any of conditions (1) to (3) is met during the week following the previous dose, the next scheduled increase in dosage is replaced with a dose of the conjugate in an amount that is the same or decreased, e.g., by 0.5 mg, relative to the dose of the previous week. The titration may be re-started at the investigator's discretion should any of conditions (1) to (3) change prior to the next dosage.
[0386] Patients are on a stable dose of > 1000 mg metformin daily for at least 3 months prior to treatment with the conjugate. Subjects undergo a routine screening evaluation up to 14 days prior to the first administration of the conjugate. Patients who have been diagnosed with Type II diabetes mellitus at least 3 months prior to screening are assessed for the following criteria:
informed consent; complete medical history; review of inclusion / exclusion criteria; survey of concomitant medications; complete physical examination; body weight; vital signs (blood pressure, temperature, pulse, respiratory rate); 12-lead ECG, urine drug screen and alcohol breath test; clinical laboratory analysis (clinical chemistry, hematology, and coagulation); urinalysis; serum pregnancy test (for pre-menopausal females only); fasting plasma glucose; FlbA lc level; fructosamine, lipid profile; total IgE level; and immunogenicity sampling.
[0387] The exendin-4(l-39) Lys40 (ε-ΑΕΕΑ-ΜΡΑ)-ΝΗ2 albumin-conjugate is administered by subcutaneous injection in the abdomen of the patient in a fasting state in the early morning. [0388] All publications, patents and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications can be made thereto without departing from the spirit or scope of the appended claims.

Claims

CLAIMS What is claimed is:
1. A method of treating a human subject having a fasting blood glucose level characteristic of the presence of diabetes, said method comprising:
(a) determining a value of the fasting blood glucose level of said subject, by a method comprising measuring said fasting blood glucose level, wherein said determining is performed by said subject;
(b) within 24 hours of said determining step, administering to the subject an amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity; wherein said administering is performed by said subject using a flexible dosage device, and wherein said administering is the first administering of said compound that occurs subsequent to said determining step;
(c) after said administering step, repeating steps (a) and (b) for a plurality of cycles, wherein each occurrence of said administering step in a cycle is with said amount being increased relative to the amount of said compound administered in the most recent cycle of said determining and administering steps, unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject; wherein if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
2. The method of claim 1 , wherein said fasting blood glucose level characteristic of the presence of diabetes is a level of greater than 125 mg/dL.
3. A method of treating a human subject having a fasting blood glucose level characteristic of the presence of pre-diabetes, said method comprising:
(a) determining a value of the fasting blood glucose level of said subject, by a method comprising measuring said fasting blood glucose level, wherein said determining is performed by said subject;
(b) within 24 hours of said determining step, administering to the subject an amount of a compound that is (1) an insulinotropic peptide; or (2) a modified insulinotropic peptide having insulinotropic activity; wherein said administering is performed by said subject using a flexible dosage device, and wherein said administering is the first administering of said compound that occurs subsequent to said determining step;
(c) after said administering step, repeating steps (a) and (b) for a plurality of cycles, wherein each occurrence of said administering step in a cycle is with said amount being increased relative to the amount of said compound administered in the most recent cycle of said determining and administering steps, unless one or more of the following occurs:
(i) the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below said fasting blood glucose level characteristic of the presence of pre-diabetes;
(ii) the subject experiences nausea or vomiting intolerable to the subject; and
(iii) the increased amount is too large practically to administer to the subject; wherein if the subject experiences one or more of said (i) - (iii) above, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same or decreased relative to the amount of said compound most recently administered to said subject.
4. The method of claim 3, wherein said fasting blood glucose level characteristic of the presence of pre-diabetes is a level in the range of 100 mg/dL to 125 mg/dL.
5. The method of any one of claims 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 80 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
6. The method of any one of claims 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 75 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
7. The method of any one of claims 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be below 70 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
8. The method of any one of claims 1 to 4, wherein if during step (c), the subject experiences a condition of diarrhea or dyspepsia intolerable to the subject, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is decreased relative to the amount of said compound most recently administered to said subject.
9. The method of claim 1 or 2, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 70 to 125 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
10. The method of claim 1 or 2, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 80 to 125 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
11. The method of any one of claims 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 70 to 99 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
12. The method of any one of claims 1 to 4, wherein if the subject experiences (i), and the value of the fasting blood glucose level of the subject is determined in an occurrence of said determining step to be in the range of 80 to 99 mg/dL, the next occurrence of said administering step comprises administering to the subject an amount of said compound that is the same as the amount of said compound most recently administered to said subject.
13. The method of any one of claims 1 to 12, wherein said value of the fasting blood glucose level of the subject is an average of a plurality of measures of the fasting blood glucose level of said subject over a period of time, wherein said plurality of measures comprises said measure obtained in said measuring step.
14. The method of any one of claims 1 to 12, wherein said value of the fasting blood glucose level of the subject is the measure of the fasting blood glucose level of said subject obtained in said measuring step.
15. The method of any one of claims 1 to 14, wherein said administering of step (b) is performed within three hours of determining the fasting blood glucose level of the subject in step (a).
16. The method of any one of claims 1 to 15, wherein each cycle of (c) comprises performing said determining and administering steps once every 1, 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, or 13 days, or once a week, or once every 2, 3, or 4 weeks, or once a month.
17. The method of any one of claims 1 to 16, wherein each cycle of step (c) comprises administering said dose within three hours of determining the fasting blood glucose level of the subject.
18. The method of any one of claims 1 to 17, wherein said repeating of step (c) comprises decreasing said dose relative to the most recently administered dose.
19. The method of any one of claims 1 to 18, wherein said repeating of step (c) comprises repeating said determining and administering steps for at least 10 cycles.
20. The method of any one of claims 1 to 19, wherein the modified insulinotropic peptide has an increased half-life relative to the non-modified insulinotropic peptide.
21. The method of any one of claims 1 to 20, wherein the modified insulinotropic peptide is an insulinotropic peptide conjugate.
22. The method of any one of claims 1 to 21, wherein the insulinotropic peptide is selected from the group consisting of a glucagon-like -peptide 1 (GLP-1) peptide, an exendin peptide, and analogs thereof.
23. The method of claim 22, wherein the insulinotropic peptide is exendin-4 or an analog thereof.
24. The method of any one of claims 1 to 23, wherein the modified insulinotropic peptide is a conjugate of albumin and an exendin peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence.
25. The method of any one of claims 1 to 24, wherein said administering the modified insulinotropic peptide comprises administering a pharmaceutical formulation comprising: a conjugate of albumin and an exendin peptide, said exendin peptide comprising a sequence which has not more than 3 amino acid substitutions, deletions, or additions relative to the native exendin-4 sequence, said conjugate being at a concentration of about 1 mg/ml to about 100 mg/ml; optionally a buffer; a tonicity modifier, wherein the tonicity modifier is at a concentration of at least 1 mM; a stabilizer; a surfactant, and optionally a preservative, wherein said formulation has a pH from about 4 to about 8.
26. The method of claim 25, wherein the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
27. The method of claim 25, wherein the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5.
28. The method of claim 25, wherein the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 160 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
29. The method of claim 25, wherein the buffer is at a concentration of no more than 0.05 M, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 15 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
30. The method of claim 25, wherein the buffer is L-histidine at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5.
31. The method of claim 25, wherein the buffer is L-histidine at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
32. The method of claim 25, wherein the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
33. The method of claim 25, wherein the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.5.
34. The method of claim 25, wherein the buffer is sodium succinate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 6.0.
35. The method of claim 25, wherein the buffer is sodium acetate at a concentration of 10 mM, said tonicity modifier is sodium chloride at a concentration of 150 mM, said stabilizer is sodium octanoate at a concentration of 5 mM, said surfactant is poloxamer 188 pluronic F68 at a concentration of 0.1% (w/v), and wherein said formulation has a pH of about 5.0.
36. The method of any one of claims 24 to 35, wherein the conjugate comprises albumin cysteine 34 thiol covalently linked to a [2-[2-[2-maleimidopropionamido(ethoxy)ethoxy]acetic acid linker covalently linked to the epsilon amino of a lysine of said exendin peptide.
37. The method of claim 36, wherein said lysine has been added to the native exendin-4 sequence.
38. The method of claim 36, wherein said lysine has been added to the carboxy terminus of the native exendin-4 sequence.
39. The method of claim 36, wherein the con ugate is according to the following:
Figure imgf000134_0001
(SEQ ID NO: 31)
wherein X is S, O, or NH of an amino acid of albumin.
40. The method of claim 39, wherein X is cysteine 34 thiol of albumin.
41. The method of any one of claims 24 to 40, wherein the albumin is human serum albumin.
42. The method of any one of claims 24 to 40, wherein the albumin is recombinant human albumin.
43. The method of any one of claims 24 to 42, wherein the conjugate comprises recombinant human serum albumin cysteine 34 thiol covalently linked to a [2- [2- [2
maleimidopropionamido(ethoxy)ethoxy] acetic acid linker covalently linked to the epsilon amino of the carboxy terminal lysine of exendin-4(l-39)Lys40-NH2.
44. The method of any one of claims 24 to 43, wherein said conjugate is at a concentration from about 1 mg/ml to about 50 mg/ml.
45. The method of any one of claims 24 to 43, wherein said conjugate is at a concentration from about 1 mg/ml to about 15 mg/ml.
46. The method of any one of claims 24 to 43, wherein said conjugate is at a concentration from about 1 mg/ml to about 10 mg/ml.
47. The method of any one of claims 24 to 43, wherein said conjugate is at a concentration of about 10 mg/ml.
48. The method of any one of claims 24 to 43, wherein said conjugate is at a concentration of about 20 mg/ml.
49. The method of any one of claims 24 to 48, wherein said dose of step (b) is at least about 0.5 mg of the conjugate.
50. The method of any one of claims 24 to 48, wherein said dose of step (b) is about 1.5 mg of the conjugate.
51. The method of any one of claims 24 to 50, wherein each cycle of step (c) comprises administering a dose of the conjugate which is about 0.1, 0.2, 0.3, 0.4 or 0.5 mg greater than the most recent prior administered dose, provided, however, that said dose does not exceed about 5.0 mg of the conjugate.
52. The method of any one of claims 1 to 51 , wherein said administering comprises subcutaneously administering the pharmaceutical formulation to the subject.
53. The method of any one of claims 1 to 52, wherein said administering comprises administering the pharmaceutical formulation to the subject with a pen-type delivery apparatus.
54. The method of claim 53, wherein said pen-type delivery apparatus comprises multiple doses of the pharmaceutical formulation.
55. The method of any one of claims 1 to 54, wherein the subject is a human.
56. The method of any one of claims 1 to 55, wherein the subject has been on a stable dose of metformin of > 1000 mg metformin daily for at least 3 months.
PCT/US2011/027315 2010-03-05 2011-03-04 Methods of administering insulinotropic peptides WO2011109787A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US31120810P 2010-03-05 2010-03-05
US61/311,208 2010-03-05

Publications (1)

Publication Number Publication Date
WO2011109787A1 true WO2011109787A1 (en) 2011-09-09

Family

ID=44542610

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/027315 WO2011109787A1 (en) 2010-03-05 2011-03-04 Methods of administering insulinotropic peptides

Country Status (1)

Country Link
WO (1) WO2011109787A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013133667A1 (en) * 2012-03-09 2013-09-12 Hanmi Science Co., Ltd. Pharmaceutical composition for the prevention or treatment of non-alcoholic fatty liver disease
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
US9278123B2 (en) 2010-12-16 2016-03-08 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl)amino)caprylic acid
EP3034090A4 (en) * 2013-08-13 2017-08-09 Shanghai Benemae Pharmaceutical Corporation Stable insulin secretagogue peptide hydro-injection pharmaceutical composition
WO2018094404A1 (en) 2016-11-21 2018-05-24 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
US9993430B2 (en) 2012-06-20 2018-06-12 Novo Nordisk A/S Tablet formulation comprising semaglutide and a delivery agent
US10335369B2 (en) 2012-03-22 2019-07-02 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
US10933120B2 (en) 2012-03-22 2021-03-02 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
US10993991B2 (en) 2015-05-22 2021-05-04 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with exendin (9-39)
CN112912100A (en) * 2018-10-26 2021-06-04 诺和诺德股份有限公司 Stable semaglutide compositions and uses thereof
US11034746B2 (en) 2011-04-12 2021-06-15 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US11116820B2 (en) 2016-03-04 2021-09-14 Eiger Biopharmaceuticals, Inc. Treatment of hyperinsulinemic hypoglycemia with exendin-4 derivatives
US11123296B2 (en) 2012-03-22 2021-09-21 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
US11318191B2 (en) 2020-02-18 2022-05-03 Novo Nordisk A/S GLP-1 compositions and uses thereof
WO2022157747A3 (en) * 2021-01-25 2022-09-01 Mylan Ireland Limited Pharmaceutical glp peptide compositions and methods of preparation thereof
US11752198B2 (en) 2017-08-24 2023-09-12 Novo Nordisk A/S GLP-1 compositions and uses thereof
US11833248B2 (en) 2018-02-02 2023-12-05 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl)amino)caprylic acid

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005027978A2 (en) * 2003-09-19 2005-03-31 Novo Nordisk A/S Albumin-binding derivatives of therapeutic peptides
WO2007053946A1 (en) * 2005-11-09 2007-05-18 Conjuchem Biotechnologies Inc. Method of treating diabetes and/or obesity with reduced nausea side effects using an insulinotropic peptide conjugated to albumin
WO2009075859A2 (en) * 2007-12-11 2009-06-18 Conjuchem Biotechnologies Inc. Formulation of insulinotropic peptide conjugates

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005027978A2 (en) * 2003-09-19 2005-03-31 Novo Nordisk A/S Albumin-binding derivatives of therapeutic peptides
WO2007053946A1 (en) * 2005-11-09 2007-05-18 Conjuchem Biotechnologies Inc. Method of treating diabetes and/or obesity with reduced nausea side effects using an insulinotropic peptide conjugated to albumin
WO2009075859A2 (en) * 2007-12-11 2009-06-18 Conjuchem Biotechnologies Inc. Formulation of insulinotropic peptide conjugates

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10960052B2 (en) 2010-12-16 2021-03-30 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl) amino) caprylic acid
US9278123B2 (en) 2010-12-16 2016-03-08 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl)amino)caprylic acid
US11382957B2 (en) 2010-12-16 2022-07-12 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl)amino)caprylic acid
US10086047B2 (en) 2010-12-16 2018-10-02 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl)amino)caprylic acid
US11034746B2 (en) 2011-04-12 2021-06-15 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US11117947B2 (en) 2011-04-12 2021-09-14 Novo Nordisk A/S Double-acylated GLP-1 derivatives
WO2013133667A1 (en) * 2012-03-09 2013-09-12 Hanmi Science Co., Ltd. Pharmaceutical composition for the prevention or treatment of non-alcoholic fatty liver disease
US11759502B2 (en) 2012-03-22 2023-09-19 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
US11759503B2 (en) 2012-03-22 2023-09-19 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
US10933120B2 (en) 2012-03-22 2021-03-02 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
US10335369B2 (en) 2012-03-22 2019-07-02 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
US11759501B2 (en) 2012-03-22 2023-09-19 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
US11123296B2 (en) 2012-03-22 2021-09-21 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
US9993430B2 (en) 2012-06-20 2018-06-12 Novo Nordisk A/S Tablet formulation comprising semaglutide and a delivery agent
US11033499B2 (en) 2012-06-20 2021-06-15 Novo Nordisk A/S Tablet formulation comprising a GLP-1 peptide and a delivery agent
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
EP3034090A4 (en) * 2013-08-13 2017-08-09 Shanghai Benemae Pharmaceutical Corporation Stable insulin secretagogue peptide hydro-injection pharmaceutical composition
US11622995B2 (en) 2015-05-22 2023-04-11 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with GLP-1 antagonists
US11617782B2 (en) 2015-05-22 2023-04-04 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with exendin (9-39)
US10993991B2 (en) 2015-05-22 2021-05-04 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with exendin (9-39)
US10993992B2 (en) 2015-05-22 2021-05-04 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with GLP-1 antagonists
US11116820B2 (en) 2016-03-04 2021-09-14 Eiger Biopharmaceuticals, Inc. Treatment of hyperinsulinemic hypoglycemia with exendin-4 derivatives
US11020484B2 (en) 2016-11-21 2021-06-01 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
AU2017361539B2 (en) * 2016-11-21 2023-06-29 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
US11738086B2 (en) 2016-11-21 2023-08-29 Eiger Biopharmaceuticals, Inc. Methods of using buffered formulations of exendin (9-39)
EP3541366A4 (en) * 2016-11-21 2020-08-05 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
WO2018094404A1 (en) 2016-11-21 2018-05-24 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
US11752198B2 (en) 2017-08-24 2023-09-12 Novo Nordisk A/S GLP-1 compositions and uses thereof
US11833248B2 (en) 2018-02-02 2023-12-05 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl)amino)caprylic acid
CN112912100A (en) * 2018-10-26 2021-06-04 诺和诺德股份有限公司 Stable semaglutide compositions and uses thereof
US11318191B2 (en) 2020-02-18 2022-05-03 Novo Nordisk A/S GLP-1 compositions and uses thereof
WO2022157747A3 (en) * 2021-01-25 2022-09-01 Mylan Ireland Limited Pharmaceutical glp peptide compositions and methods of preparation thereof

Similar Documents

Publication Publication Date Title
US20090186819A1 (en) Formulation of insulinotropic peptide conjugates
WO2011109787A1 (en) Methods of administering insulinotropic peptides
WO2011109784A1 (en) Formulation of insulinotropic peptide conjugates
JP6685343B2 (en) Liquid formulation of long-acting insulinotropic peptide conjugate
JP6686070B2 (en) Liquid formulation of long-acting insulin and insulinotropic peptide
RU2573995C2 (en) Pharmaceutical composition containing glp-1 agonist and methionine
JP7461997B2 (en) ELP Fusion Proteins for Controlled and Sustained Release
JP4353544B2 (en) Amylin agonist peptide formulation
ZA200406626B (en) Method for administering glp-1 molecules
US20090202494A1 (en) Combined use of glp-1 agonists and gastrin for regulating blood glucose levels
AU2011348202A9 (en) Methods for treating metabolic disorders and obesity with GIP and GLP-1 receptor-active glucagon-based peptides
EP1049486A1 (en) Glp-1 formulations
JP2011506442A5 (en)
WO2008056726A1 (en) Glp-1 derivative and use thereof
US20130005652A1 (en) Methods for administering long-lasting hypoglycemic agents
TWI353250B (en) Glp-1 pharmaceutical compositions
JP2016505627A (en) Therapeutic agents, compositions, and methods for glycemic control
US20020123466A1 (en) GLP-1 formulations
MXPA01005648A (en) Shelf-stable formulation of glucagon-like peptide-1

Legal Events

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

Ref document number: 11751479

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11751479

Country of ref document: EP

Kind code of ref document: A1