US20150290328A1 - Formulations of active agents for sustained release - Google Patents

Formulations of active agents for sustained release Download PDF

Info

Publication number
US20150290328A1
US20150290328A1 US14/646,273 US201314646273A US2015290328A1 US 20150290328 A1 US20150290328 A1 US 20150290328A1 US 201314646273 A US201314646273 A US 201314646273A US 2015290328 A1 US2015290328 A1 US 2015290328A1
Authority
US
United States
Prior art keywords
formulation
matrix
amino acid
acid sequence
active agent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/646,273
Inventor
Susan Arnold
Christopher Prior
Lynne Georgopoulos
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Phasebio Pharmaceuticals Inc
Original Assignee
Phasebio Pharmaceuticals Inc
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 Phasebio Pharmaceuticals Inc filed Critical Phasebio Pharmaceuticals Inc
Priority to US14/646,273 priority Critical patent/US20150290328A1/en
Assigned to PHASEBIO PHARMACEUTICALS, INC. reassignment PHASEBIO PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PRIOR, CHRISTOPHER, ARNOLD, SUSAN, GEORGOPOULOS, Lynne
Assigned to PHASEBIO PHARMACEUTICALS, INC. reassignment PHASEBIO PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PRIOR, CHRISTOPHER, ARNOLD, SUSAN, GEORGOPOULOS, Lynne
Publication of US20150290328A1 publication Critical patent/US20150290328A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2278Vasoactive intestinal peptide [VIP]; Related peptides (e.g. Exendin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/26Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • A61K47/48292
    • 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
    • A61K47/6435Drug-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 the peptide or protein in the drug conjugate being a connective tissue peptide, e.g. collagen, fibronectin or gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to pharmaceutical formulations for sustained release when administered at cold temperatures, and methods for delivering a treatment regimen with the sustained release formulations.
  • peptide and small molecule drugs are often limited by the half-life of such drugs in the circulation, as well as difficulties in obtaining substantially constant plasma levels.
  • the incretin GLP-1 must be administered at relatively high doses to counter its short half-life in the circulation, and these high doses are associated with nausea, among other things. Murphy and Bloom, Nonpeptidic glucagon - like peptide 1 receptor agonists: A magic bullet for diabetes? PNAS 104 (3):689-690 (2007).
  • the peptide agent vasoactive intestinal peptide (VIP) exhibits a half-life, in some estimates, of less than one minute, making this agent impractical for pharmaceutical use.
  • Domschke et al. Vasoactive intestinal peptide in man: pharmacokinetics, metabolic and circulatory effects.
  • a short plasma half life for peptide drugs is often due to fast renal clearance as well as to enzymatic degradation during systemic circulation.
  • the present invention provides pharmaceutical formulations for sustained release, and methods for delivering a treatment regimen with the sustained release formulations.
  • the invention thereby provides improved pharmacokinetics for peptide and small molecule drugs.
  • the invention provides a sustained release pharmaceutical formulation.
  • the formulation comprises a therapeutic agent for systemic administration, where the therapeutic agent comprises an active agent and an amino acid sequence capable of forming a reversible matrix at the body temperature of a subject.
  • the reversible matrix is formed from hydrogen bonds (e.g., intra- and/or intermolecular hydrogen bonds) as well as from hydrophobic contributions.
  • the formulation further comprises one or more pharmaceutically acceptable excipients and/or diluents inducing the formation of the matrix upon administration.
  • the matrix provides for a slow absorption to the circulation from an injection site.
  • the sustained release, or slow absorption from the injection site is due to a slow reversal of the matrix as the concentration dissipates at the injection site.
  • the formulation confers long half-life and improved stability.
  • a unique combination of slow absorption and long half-life is achieved leading to a desirable PK profile with a shallow peak to trough ratio and/or long Tmax.
  • these benefits can be provided by administering cold formulations (e.g. 2-15° C., or 2-1.0° C., or 2-5° C.) of the therapeutic agent.
  • the amino acid sequence is an Elastin-Like-Protein (ELP) sequence.
  • ELP sequence comprises or consists of structural peptide units or sequences that are related to, or mimics of, the elastin protein.
  • the amino acid sequence may exhibit a visible and reversible inverse phase transition with the selected formulation. That is, the amino acid sequence may be structurally disordered and highly soluble in the formulation below a transition temperature (Tt), but exhibit a sharp (2-3° C. range) disorder-to-order phase transition when the temperature of the formulation is raised above the Tt.
  • Tt transition temperature
  • amino acid polymer In addition to temperature, length of the amino acid polymer, amino acid composition, ionic strength, pH, pressure, selected solvents, presence of organic solutes, temperature, and protein concentration may also affect the transition properties, and these may be tailored for the desired absorption profile.
  • Other exemplary sequences or structures for the amino acid sequence forming the matrix are disclosed herein.
  • the active agent for systemic administration is a protein or peptide, which may have a short circulatory half-fife, such as from about 30 seconds to about 1 hour, to about 2 hours, or to about 5 hours. In some embodiments, the protein or peptide has a circulatory half-life of from 30 seconds to about 10 hours.
  • the therapeutic agent may be a recombinant fusion protein between the protein active agent and the amino acid sequence capable of forming the matrix.
  • Exemplary peptide active agents include GLP-1 receptor agonists (e.g., GLP-1 or derivative thereof), glucagon receptor agonists (e.g. glucagon, oxyntomodulin or derivatives thereof), VPAC2 selective agonists (e.g.
  • vasoactive intestinal peptide VIP or a derivative thereof
  • GIP receptor agonists e.g. glucose-dependent insulinotropic peptide (GIP) or a derivative thereof
  • insulin or a derivative thereof e.g. glucose-dependent insulinotropic peptide (GIP) or a derivative thereof
  • the protein active agent is a clotting factor, such as Factor VII, Factor VIII, or Factor IX.
  • Other protein and small molecule drugs for delivery in accordance with the invention are disclosed herein.
  • the invention provides a method for delivering a sustained release regimen of an active agent.
  • the method comprises administering the formulation described herein to a subject in need, wherein the formulation is administered from about 1 to about 8 times per month.
  • the formulation is administered about weekly, and may be administered subcutaneously or intramuscularly (for example).
  • the site of administration is not a pathological site, that is, the therapeutic agent is not administered directly to the intended site of action.
  • FIG. 1 shows the phase transition (as shown by an increase in turbidity) of an ELP1 protein, induced by a change in temperature to 37° C. or above. This property provides for a slow absorption from an injection site.
  • FIG. 2 shows the phase transition (as shown by an increase in turbidity) of an ELP4 protein, induced by a change in temperature to 25° C. or above. This property provides for a depot-like delivery.
  • FIG. 3 illustrates, without wishing to be bound by theory, a potential mechanism for the observed transition, in which a water shell is excluded under certain conditions, allowing for hydrogen bonds to form.
  • FIG. 4 shows that the ELP4 series transitions at 37° C. at a protein concentration of less than about 0.01 mg/ml, allowing for sustained release formulations of low protein concentration, for example, at the injection site
  • FIG. 5 shows that the ELP1 series transitions at just below 37° C. at relatively high protein concentration of about 10 mg/ml or more, allowing for sustained release formulations with relatively high amounts of active agents.
  • FIG. 6 shows a summary of pharmacokinetic parameters for Glp-1/ELP1-120 (also referred to herein as PB1023 or Glymera) after SC administration of 0.3, 0.6, 0.9 and 1.35 mg/kg to adult subjects with type 2 diabetes mellitus.
  • FIG. 7 shows the mean serum concentrations of Glp-1/ELP1-120 (also referred to herein as PB1023 or Glymera) after s.c. administration on day 0 of 0.3, 0.6, 0.9 and 1.35 mg/kg to adult subjects with type 2 diabetes mellitus (semi-logarithmic axes).
  • FIG. 8 shows the type 2 diabetes mellitus: Glymera program overview. pharmacokinetics crossover study. Mean serum concentrations of Glymera following s.c. administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with type 2 diabetes mellitus are shown (semi-logarithmic axes).
  • FIG. 9 shows a summary of pharmacokinetic parameters for Glymera after s.c. administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with type 2 diabetes mellitus.
  • FIG. 10 shows a summary of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as 50 mg/mL and 100 mg/mL formulations at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM.
  • FIG. 11 shows a statistical comparison of pharmacokinetic parameters for PB1023 alter SC administration of 90 mg as 50 mg/mL and 100 mg/mL formulations at room temperature and 100 mg/mL, cold (2° to 8° C.) to adult subjects with T2DM.
  • FIG. 12 shows the mean ⁇ standard deviation serum concentrations of PB1023 after SC administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with T2DM—linear axes.
  • FIG. 13 shows the mean ⁇ standard deviation serum concentrations of PB1023 after SC administration of 90 mg as 50 mg/mL, and 100 mg/mL formulations to adult subjects with T2DM—semi-logarithmic axes.
  • FIG. 14 shows a summary of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as 50 mg/mL, and 100 mg/mL, formulations to adult subjects with T2DM.
  • FIG. 15 shows a statistical comparison of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with T2DM.
  • FIG. 16 shows the mean ⁇ standard deviation serum concentrations of PB1023 after SC administration of 90 mg as the 100 mg/mL formulation at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM linear axes.
  • FIG. 17 shows the mean ⁇ standard deviation serum concentrations of PB1023 after SC administration of 90 mg as the 100 mg/mL, formulation at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM semi-logarithmic axes.
  • FIG. 18 shows a summary of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as the 100 mg/mL formulation at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM.
  • FIG. 19 shows a statistical comparison of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as the 100 mg/mL formulation at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM.
  • FIG. 20A shows the human proinsulin sequence.
  • the proinsulin sequence consists of the B and A chains linked with the C peptide.
  • the C peptide is removed to form mature insulin following enzymatic cleavage at the two adjacent dibasic sites (underlined in italics).
  • FIG. 20B shows the amino acid sequence of a proinsulin ELP1-120 fusion protein.
  • the proinsulin sequence (underlined) is fused to the ELP1-120 sequence.
  • the amino acid sequence optionally includes an initiation methionine residue at the N terminus.
  • FIG. 21 shows the amino acid sequence of a GLP-1/ELP fusion protein as described herein.
  • the present invention provides pharmaceutical formulations for sustained release, and methods for delivering a treatment regimen with the sustained release formulations.
  • the invention thereby provides improved pharmacokinetics for peptide and small molecule drugs, including a relatively flat PK profile with a low ratio of peak to trough, and/or a long Tmax.
  • the PK profile can be maintained with a relatively infrequent administration schedule, such as from one to eight injections per month in some embodiments.
  • the invention provides a sustained release pharmaceutical formulation.
  • the formulation comprises a therapeutic agent for systemic administration, where the therapeutic agent comprises an active agent and an amino acid sequence capable of forming a matrix at the body temperature of a subject.
  • the reversible matrix is formed from hydrogen bonds (e.g., intra- and/or intermolecular hydrogen bonds) as well as from hydrophobic contributions.
  • the formulation further comprises one or more pharmaceutically acceptable excipients and/or diluents inducing the formation of the matrix upon administration.
  • the matrix provides for a slow absorption to the circulation from an injection site, and without being bound by theory, this slow absorption is due to the slow reversal of the matrix as protein concentration decreases at the injection site.
  • the slow absorption profile provides for a flat PK profile, as well as convenient and comfortable administration regimen.
  • the plasma concentration of the active agent over the course of days e.g., from 2 to about 60 days, or from about 4 to about 30 days
  • this flat PK profile is seen over a plurality of (substantially evenly spaced) administrations, such as at least 2, at least about 5, or at least about 10 administrations of the formulation.
  • the slow absorption is exhibited by a Tmax (time to maximum plasma concentration) of greater than about 5 hours, greater than about 10 hours, greater than about 20 hours, greater than about 30 hours, or greater than about 50 hours.
  • the sustained release, or slow absorption from the injection site is controlled by the amino acid sequence capable of forming a hydrogen-bonded matrix at the body temperature of the subject, as well as the components of the formulation.
  • the amino acid sequence contains structural units that farm hydrogen-bonds through protein backbone groups and/or side chain groups, and which may contribute hydrophobic interactions to matrix formation.
  • the amino acids side chains do not contain hydrogen bond donor groups, with hydrogen bonds being formed substantially through the protein backbone.
  • Exemplary amino acids include proline, alanine, valine, glycine, and isoleucine, and similar amino acids.
  • the structural units are substantially repeating structural units, so as to create a substantially repeating structural motif, and substantially repeating hydrogen-bonding capability.
  • the amino acid sequence contains at least 10%, at least 20%, at least 40%, or at least 50% proline, which may be positioned in a substantially repeating pattern.
  • a substantially repeating pattern means that at least 50% or at least 75% of the proline residues of the amino acid sequence are part of a definable structural unit.
  • the amino acid sequence contains amino acids with hydrogen-bond donor side chains, such as serine, threonine, and/or tyrosine.
  • the repeating sequence may contain from one to about four proline residues, with remaining residues independently selected from non-polar residues, such as glycine, alanine, leucine, isoleucine, and valine. Non-polar or hydrophobic residues may contribute hydrophobic interactions to the formation of the matrix.
  • amino acid sequences may form a “gel-like” state upon injection at a temperature higher than the storage temperature.
  • Exemplary sequences have repeating peptide units, and/or may be relatively unstructured at the lower temperature, and achieve a hydrogen-bonded, structured, state at the higher temperature.
  • the amino acid sequence capable of forming the matrix at body temperature is a peptide having repeating units of from four to ten amino acids.
  • the repeating unit may form one, two, or three hydrogen bonds in the formation of the matrix.
  • the amino acid sequence capable of forming the matrix at body temperature is an amino acid sequence of silk, elastin, collagen, or keratin, or mimic thereof, or an amino acid sequence disclosed in U.S. Pat. No. 6,355,776, which is hereby incorporated by reference.
  • the amino acid sequence is an Elastin-Like-Protein (ELP) sequence.
  • ELP sequence comprises or consists of structural peptide units or sequences that are related to, or mimics of, the elastin protein.
  • the ELP sequence is constructed from structural units of from three to about twenty amino acids, or in some embodiments, from four to ten amino acids, such as four, five or six amino acids.
  • the length of the individual structural units may vary or may be uniform.
  • Exemplary structural units include units defined by SEQ ID NOS: 1-12 (below), which may be employed as repeating structural units, including tandem-repeating units, or may be employed in some combination.
  • the ELP may comprise or consist essentially of structural unit(s) selected from SEQ ID NOS: 1-12, as defined below.
  • the amino acid sequence comprises or consists essentially of from about 10 to about 500 structural units, or in certain embodiments about 50 to about 200 structural units, or in certain embodiments from about 80 to about 200 structural units, or from about 80 to about 150 structural units, such as one or a combination of units defined by SEQ ID NOS: 1-12.
  • the structural units collectively may have a length of from about 50 to about 2000 amino acid residues, or from about 100 to about 800 amino acid residues, or from about 200 to about 700 amino acid residues, or from about 400 to about 600 amino acid residues.
  • the amino acid sequence may exhibit a visible and reversible inverse phase transition with the selected formulation. That is, the amino acid sequence may be structurally disordered and highly soluble in the formulation below a transition temperature (Tt), but exhibit a sharp (2-3° C. range) disorder-to-order phase transition when the temperature of the formulation is raised above the Tt.
  • Tt transition temperature
  • length of the amino acid polymer, amino acid composition, ionic strength, pH, pressure, temperature, selected solvents, presence of organic solutes, and protein concentration may also affect the transition properties, and these may be tailored in the formulation for the desired absorption profile. Absorption profile can be easily tested by determining plasma concentration or activity of the active agent over time.
  • the ELP component(s) may be formed of structural units, including but not limited to:
  • Such structural units defined by SEQ ID NOS:1-12 may firm structural repeat units, or may be used in combination to form an ELP.
  • the ELP component is formed entirely (or almost entirely) of one or a combination of (e.g., 2, 3 or 4) structural units selected from SEQ ID NOS: 1-12.
  • at least 75%, or at least 80%, or at least 90% of the ELP component is formed from one or a combination of structural units selected from SEQ ID NOS: 1-12, and which may be present as repeating units.
  • the ELP contains repeat units, including tandem repeating units, of Val-Pro-Gly-X-Gly (SEQ ID NO: 3), where X is as defined above, and where the percentage of Val-Pro-Gly-X-Gly (SEQ ID NO: 3) units taken with respect to the entire ELP component (which may comprise structural units other than VPGXG (SEQ ID NO: 3)) is greater than about 50%, or greater than about 75%, or greater than about 85%, or greater than about 95% of the ELP.
  • the ELP may contain motifs of 5 to 15 structural units (e.g. about 10 structural units) of SEQ ID NO: 3, with the guest residue X varying among at least 2 or at least 3 of the units in the motif.
  • the guest residues may be independently selected, such as from non-polar or hydrophobic residues, such as the amino acids V, I, L, A, G, and W (and may be selected so as to retain a desired inverse phase transition property).
  • the ELP may form a 13-turn structure.
  • Exemplary peptide sequences suitable tier creating a ⁇ -turn structure are described in International Patent Application PCT/US96/05186, which is hereby incorporated by reference in its entirety.
  • the fourth residue (X) in the sequence VPGXG (SEQ ID NO: 3) can be altered without eliminating the formation of a ⁇ -turn.
  • ELPk designates a particular ELP repeat unit
  • bracketed capital letters are single letter amino acid codes and their corresponding subscripts designate the relative ratio of each guest residue X in the structural units (where applicable)
  • n describes the total length of the [[P in number of the structural repeats.
  • ELP1 [V5A2G3-10] designates an ELP component containing 10 repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is valine, alanine, and glycine at a relative ratio of about 5:2:3;
  • ELP1 [K1V2F1-4] designates an ELP component containing 4 repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is lysine, valine, and phenylalanine at a relative ratio of about 1:2:1;
  • ELP1 [K1V7F1-9] designates a polypeptide containing 9 repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is lysine, valine, and phenylalanine at a relative ratio of about 1:7:1;
  • ELP1 [V-5] designates a polypeptide containing 5 repeating units of the pentapeptide
  • the Tt is a function of the hydrophobicity of the guest residue.
  • ELPs can be synthesized that exhibit an inverse transition over a broad range.
  • the Tt at a given ELP length may be decreased by incorporating a larger fraction of hydrophobic guest residues in the ELP sequence.
  • suitable hydrophobic guest residues include valine, leucine, isoleucine, phenylalanine, tryptophan and methionine. Tyrosine, which is moderately hydrophobic, may also be used.
  • the It may be increased by incorporating residues, such as those selected from: glutamic acid, cysteine, lysine, aspartate, alanine, asparagine, serine, threonine, glycine, arginine, and glutamine.
  • residues such as those selected from: glutamic acid, cysteine, lysine, aspartate, alanine, asparagine, serine, threonine, glycine, arginine, and glutamine.
  • the hydrophobicity scale disclosed in PCT/US96/05186 provides one means far predicting the approximate Tt of a specific ELP sequence.
  • the ELP in some embodiments is selected or designed to provide a Tt ranging from about 10 to about 37° C. at formulation conditions, such as from about 20 to about 37° C., or from about 25 to about 37° C.
  • the transition temperature at physiological conditions e.g., 0.9% saline
  • the amino acid sequence capable of farming the hydrogen-bonded matrix at body temperature comprises [VPGXG] 90 , where each X is selected from V, G, and A, and wherein the ratio of V:G:A may be about 5:3:2.
  • the amino acid sequence capable of forming the hydrogen-bonded matrix at body temperature may comprise [VPGXG] 120 , where each X is selected from V, G, and A, and wherein the ratio of V:G:A may be about 5:3:2.
  • 120 structural units of this ELP can provide a transition temperature at about 37° C. with about 5 to 15 mg/ml. (e.g., about 10 mg/ml) of protein. At concentrations of about 50 to about 100 mg/mL the phase transition temperature is about 35.5 degrees centigrade (just below body temperature), which allows for peripheral body temperature to be just less than 37° C.
  • the amino acid sequence capable of forming the matrix at body temperature comprises [VPGVG] 90 , or [VPGVG] 120 .
  • 120 structural units of this ELP can provide a transition temperature at about 37° C. with about 0.005 to about 0.05 mg/ml (e.g., about 0.01 mg/ml) of protein.
  • Elastin-like-peptide (ELP) protein polymers and recombinant fusion proteins can be prepared as described in U.S. Patent Publication No. 2010/0022455, which is hereby incorporated by reference.
  • the amino acid sequence capable of forming the matrix at body temperature may include a random coil or non-globular extended structure.
  • the amino acid sequence capable of forming the matrix at body temperature may comprise an amino acid sequence disclosed in U.S. Patent Publication No. 2008/0286808, WIPO Patent Publication No. 2008/155134, and U.S. Patent Publication No. 2011/0123487, each of which is hereby incorporated by reference.
  • the amino acid sequence comprises an unstructured recombinant polymer of at least 40 amino acids.
  • the unstructured polymer may be defined where the sum of glycine (G), aspartate (D), alanine (A), serine (S), threonine (T), glutamate (E) and proline (P) residues contained in the unstructured polymer, constitutes more than about 80% of the total amino acids.
  • at least 50% of the amino acids are devoid of secondary structure as determined by the Chou-Fasman algorithm.
  • the unstructured polymer may comprise more than about 100, 150, 200 or more contiguous amino acids.
  • the amino acid sequence forms a random coil domain.
  • a polypeptide or amino acid polymer having or forming “random coil conformation” substantially lacks a defined secondary and tertiary structure.
  • the intended subject is human, and the body temperature is about 37° C., and thus the therapeutic agent is designed to provide a sustained release at this temperature.
  • a slow release into the circulation with reversal of hydrogen bonding and/or hydrophobic interactions is driven by a drop in concentration as the product diffuses at the injection site, even though body temperature remains constant.
  • the subject is a non-human mammal, and the therapeutic agent is designed to exhibit a sustained release at the body temperature of the mammal, which may be from about 30 to about 40° C. in some embodiments, such as for certain domesticated pets (e.g., dog or cat) or livestock (e.g., cow, horse, sheep, or pig).
  • the Tt is higher than the storage conditions of the formulation.
  • the storage conditions may be from about 2 to about 22° C., including about 2 to about 3° C., about 2 to about 4° C., about 2 to about 5° C., about 2 to about 6° C., about 2 to about 7° C., about 2 to about 8° C., about 2 to about 10° C., about 2 to about 12° C., about 2 to about 14° C., about 2 to about 16° C., about 10 to about 25° C., or from 15 to 22° C., such that the therapeutic agent remains in solution for injection and exhibits the desired pharmacokinetic behavior.
  • the formulation may be administered at the about storage temperature.
  • the therapeutic agent is generally for “systemic delivery,” meaning that the agent is not delivered locally to a pathological site or a site of action. Instead, the agent is absorbed into the bloodstream from the injection site, where the agent acts systemically or is transported to a site of action via the circulation.
  • the active agent is a protein or peptide, which may have a short circulatory half-life, such as from about 30 seconds to about 1 hour.
  • the therapeutic agent may be a recombinant fusion protein between the protein active agent and the amino acid sequence capable of forming the hydrogen-bonded matrix at the body temperature of the subject.
  • Exemplary peptide active agents include GIP receptor agonists such as glucose-dependent insulinotropic peptide (GIP) or a derivative thereof.
  • GIP glucose-dependent insulinotropic peptide
  • Further exemplary peptide active agents include GLP1 receptor agonists such as GLP-1 or derivative thereof (including GLP1 7-36 or GLP1 7-37), or exendin or derivative thereof.
  • GLP1 7-36 has the following amino acid sequence: HAEGTFTSDVSSYLEGQAAKEFIAWLVKGR.
  • Exendin-4 has the following amino acid sequence: HGEGTFTSDLSKQMEEEAVRLFEWLKNGGPSSGAPPPS.
  • the protein or peptide agent is a glucagon receptor agonist (including glucagon, oxyntomodulin or a derivative thereof).
  • Oxyntomodulin has the amino acid sequence HSQGTFTSDYSKYLDSRRAQDFVQWLMNTKRNKNNIA.
  • Glucagon has the amino acid sequence HSQGTFTSDYSKYLDSRRAQDFVQWLMNT.
  • GLP-2 has the amino acid sequence HADGSFSDEMNTILDNLAARDFINWLIQTKITD.
  • the amino acid sequence of GIP is YAEGTFISDYSIAMDKIRQQDFVNWLLAQ.
  • the GLP-1 receptor agonist is a dual agonist having an amino acid sequence described in US 2011/0257092, which is hereby incorporated by reference in its entirety.
  • Other dual or multi receptor agonists are described in US 2011/016602 and US 2010/00190701, each of which is hereby incorporated by reference, in particular with regard to the structures and sequences of GLP-1 receptor co-agonists described therein.
  • GLP-1 receptor co-agonists can be found in Pocai A et al., Glucagon-Like Peptide 1/Glucagon Receptor Dual Agonism Reverses Obesity in Mice, Diabetes 58:2258-2266 (2009) and Patterson J T et al., Functional association of the N-terminal residues with the central region in glucagon-related peptides, J. Pept. Sci. 17:659-666 (2011), each of which are hereby incorporated by reference in their entirety.
  • the invention provides for a co-formulation of any two of a GLP1 receptor agonist, a glucagon receptor agonist, and a GIP receptor agonist.
  • the protein or peptide agent is a VPAC2 selective agonist, such as vasoactive intestinal peptide (VIP) or a derivative thereof.
  • VPAC2 selective agonist such as vasoactive intestinal peptide (VIP) or a derivative thereof.
  • the protein active agent is a clotting factor, such as Factor VII, Factor VIII, or Factor IX, or in other embodiments is insulin (e.g., single chain insulin or an A chain or a B chain fusion protein, as described in U.S. Provisional Application No. 61/563,985, which is hereby incorporated by reference) or a monoclonal antibody or single chain antibody.
  • the active agent is as described in U.S. Patent Publication No. 2011/0123487, which is hereby incorporated by reference.
  • Exemplary therapeutic agents in accordance with the invention include GLP-1 (A8G,7-37) ELP1-120 (referred to herein as PB1023) or GLP-1 (A8G,7-37) ELP4-120 (PB1046).
  • PB1023 GLP-1 (A8G,7-37) ELP1-120
  • PB1046 GLP-1 (A8G,7-37) ELP4-120
  • the invention encompasses formulations which comprise a therapeutic agent for systemic administration, where the therapeutic agent comprises an active agent selected from one or more of insulin (by way of non-limiting example, single chain insulin or an A chain or a B chain fusion protein) and GLP-1 or derivative thereof (by way of non-limiting example, GLP1 7-36 or GLP1 7-37) and an amino acid sequence capable of forming a matrix at the body temperature of a subject (by way of non-limiting example, ELP).
  • insulin by way of non-limiting example, single chain insulin or an A chain or a B chain fusion protein
  • GLP-1 or derivative thereof by way of non-limiting example, GLP1 7-36 or GLP1 7-37
  • ELP amino acid sequence capable of forming a matrix at the body temperature of a subject
  • the present invention encompasses a formulation, or co-formulation, of GLP-1 or derivative thereof (including GLP1 7-36 or GLP1 7-37) fused to ELP and insulin (by way of non-limiting example, single chain insulin or an A chain or a B chain fusion protein) fused to ELP.
  • these formulations are administered cold (by way of non-limiting example, 2-10° C.) or at room temperature as described herein.
  • the invention encompasses doses and/or regimens such as those that do not induce substantial appetite suppression in a patient and/or those that do not induce substantial nausea in the patient, such as those described in PCT/US12/44383, which is hereby incorporated by reference.
  • the therapeutic agent is a chemical conjugate between the active agent and the amino acid sequence capable of forming the matrix at the body temperature of the subject.
  • the active agent may be a chemotherapeutic agent, such as a chemotherapeutic agent selected from methotrexate, daunomycin, mitomycin, cisplatin, vincristine, epirubicin, fluorouracil, verapamil, cyclophosphamide, cytosine arabinosidc, aminopterin, bleomycin, mitomycin C, democolcine, etoposide, mithramycin, chlorambucil, melphalan, daunoubicin, doxorubicin, tamoxifen, paclitaxel, vinblastine, camptothecin, actinomycin 13, cytarabine, and combrestatin.
  • a chemotherapeutic agent selected from methotrexate, daunomycin, mitomycin, cisplatin, vincris
  • the agent may be an immunogenic molecule, or an immunomodulator, or an anti-inflammatory agent, such as an agent described in U.S. Patent Publication No. 2009/0004104, which is hereby incorporated by reference in its entirety.
  • the agent may be an opioid molecule, such as, for example oxycodone, morphine, or codeine such as described in U.S. Provisional Application No. 61/597,898, which is hereby incorporated by reference.
  • the chemical conjugate may be through a cleavable linker, for which numerous types are known in the art. See U.S. Pat. No. 6,326,996, which is hereby incorporated by reference in its entirety.
  • the formulation comprises one or more pharmaceutically acceptable excipients and/or diluents inducing the formation of the matrix upon administration.
  • excipients include salts, and other excipients that may act to stabilize hydrogen bonding.
  • exemplary salts include alkaline earth metal salts such as sodium, potassium, and calcium.
  • Counter ions include chloride and phosphate.
  • Exemplary salts include sodium chloride, potassium chloride, magnesium chloride, calcium chloride, and potassium phosphate.
  • the protein concentration in the formulation is tailored to drive, along with the excipients, the formation of the matrix at the temperature of administration. For example, higher protein concentrations help drive the formation of the matrix, and the protein concentration needed for this purpose varies depending on the ELP series used.
  • the protein is present in the range of about 1 mg/mL, to about 200 mg/mL, or is present in the range of about 5 mg/mL, to about 125 mg/mL.
  • the therapeutic agent may be present in the range of about 10 mg/mL to about 50 mg/mL, or about 15 mg/mL to about 30 mg/mL.
  • the protein is present in the range of about 0.005 mg/mL to about 10 mg/mL, or is present in the range of about 0.01 mg/mL to about 5 mg/mL.
  • the therapeutic agent is formulated at a pH, ionic strength, and generally with excipients sufficient to drive the formation of the matrix at body temperature (e.g., 37° C., or at from 34 to 36° C. in some embodiments).
  • the therapeutic agent is generally prepared such that it does not form the matrix at storage conditions. Storage conditions are generally less than the transition temperature of the formulation, such as less than about 32° C., or less than about 30° C., or less than about 27° C., or less than about 25° C., or less than about 20° C., or less than about 15° C.
  • the formulation may be isotonic with blood or have an ionic strength that mimics physiological conditions.
  • the formulation may have an ionic strength of at least that of 25 mM Sodium Chloride, or at least that of 30 mM Sodium chloride, or at least that of 40 mM Sodium Chloride, or at least that of 50 mM Sodium Chloride, or at least that of 75 mM Sodium Chloride, or at least that of 100 mM Sodium Chloride, or at least that of 150 mM Sodium Chloride.
  • the formulation has an ionic strength less than that of 0.9% saline.
  • the formulation comprises two or more of calcium chloride, magnesium chloride, potassium chloride, potassium phosphate monobasic, sodium chloride, and sodium phosphate dibasic.
  • the liquid formulation may comprise the components listed in Table 4, Table 5, or Table 6, and can be stored refrigerated or at room temperature.
  • the formulation can be packaged in the form of pre-dosed pens or syringes for administration once per week, twice per week, or from one to eight times per month, or alternatively filled in conventional vial and the like.
  • the invention provides a sustained release pharmaceutical formulation that comprises a therapeutic agent, the therapeutic agent (e.g., a peptide or protein therapeutic agent) comprising an active agent and an amino acid sequence comprising [VPGXG] 90 , or [VPGXG] 120 , where each X is selected from V, G, and A. V, G, and A may be present at a ratio of about 5:3:2.
  • the amino acid sequence comprises [VPGXG] 90 , or [VPGXG] 120 .
  • the formulation further comprises one or more pharmaceutically acceptable excipients and/or diluents for formation of a reversible matrix from an aqueous form upon administration to a human subject.
  • the active agent in certain embodiments is GLP-1 or derivative thereof (e.g., GLP-1, A8G, 7-37), or vasoactive intestinal peptide (VIP) or a derivative thereof (e.g., having an N-terminal moiety such as a Methionine), or oxyntomodulin of a derivative thereof, or insulin or a derivative thereof.
  • GLP-1 and derivatives thereof are disclosed in U.S. Patent Publication No. 2011/0123487, which is hereby incorporated by reference.
  • VIP and derivatives thereof are disclosed in U.S. Patent Publication No. 2011/0178017, which is hereby incorporated by reference.
  • Insulin and derivatives thereof are described in U.S. Provisional Application No. 61/563,985, which is hereby incorporated by reference.
  • the therapeutic agent may be present in the range of about 0.5 mg/mL to about 200 mg/mL, or is present in the range of about 5 mg/mL to about 125 mg/ML.
  • the therapeutic agent is present in the range of about 10 mg/mL to about 50 mg/nit, or the range of about 15 mg/mL, to about 30 mg/mL.
  • the formulation may have an ionic strength of at least that of 25 mM Sodium Chloride, or at least that of 30 mg/mL sodium Chloride, or at least that of 40 mM Sodium Chloride, or at that least that of 50 mM Sodium Chloride, or at least that of 75 mM Sodium Chloride, or at least that of 100 mM Sodium Chloride.
  • the formulation may have an ionic strength less than that of about 0.9% saline.
  • the formulation comprises two or more of calcium chloride, magnesium chloride, potassium chloride, potassium phosphate monobasic, sodium chloride, and sodium phosphate dibasic.
  • the formulation may comprise the components listed in Table 4, Table 5, or Table 6.
  • formulation components for achieving the desired stability may also be employed.
  • Such components include one or more amino acids or sugar alcohol (e.g., mannitol), preservatives, and buffering agents, and such ingredients are well known in the art.
  • the invention provides a method for delivering a sustained release regimen of an active agent.
  • the method comprises administering the formulation described herein to a subject in need, wherein the formulation is administered from about 1 to about 8 times per month (e.g., about weekly).
  • the active agent may be GLP-1 or an analog thereof, and is administered in a method described in U.S. patent application Ser. No. 13/534,836, which is hereby incorporated by reference.
  • the therapeutic agent may be GLP-1 7-36 or 7-37, alternatively having Gly at position 8, fused to ELP1 (e.g., having from about 90 to about 150 ELP units).
  • the GLP-1 fusion may be used for the treatment of diabetes (type 1 or 2), metabolic disease, or obesity, for example, by administering to a patient in need.
  • the active agent is VIP or an analog thereof, and is administered in a method described in U.S. Patent Publication No. 2011/0178017, which is hereby incorporated by reference.
  • the VIP may have an additional moiety such as Methionine at the N-terminus to alter the receptor binding profile, as also described in U.S. Patent Publication No. 2011/0178017, which description is hereby incorporated by reference.
  • the VIP may be fused to ELP1 (having from about 90 to about 150 ELP units).
  • the VIP active agent finds use in a method of treating a condition selected from uncontrolled or resistant hypertension, or pulmonary arterial hypertension (PAH), and chronic obstructive pulmonary disease (COPD), among others.
  • the active agent is insulin, which finds use in methods (using the ELP-based regimens described herein) of treating diabetes, including type 1 or type 2 diabetes.
  • the formulation is administered about weekly, and may be administered subcutaneously or intramuscularly.
  • the site of administration is not a pathological site, for example, is not the intended site of action.
  • the plasma concentration of the active agent does not change by more than a factor of 10, or a factor of about 5, or a factor of about 3 over the course of a plurality of administrations, such as at least 2, at least about 5, or at least about 10 administrations of the formulation.
  • the administrations are substantially evenly spaced, such as, for example, about daily, or about once per week, or from one to about five times per month.
  • the subject is a human, but in other embodiments may be a non-human mammal, such as a domesticated pet (e.g., dog or cat), or livestock or farm animal (e.g., horse, cow, sheep, or pig).
  • a domesticated pet e.g., dog or cat
  • livestock or farm animal e.g., horse, cow, sheep, or pig
  • FIG. 1 illustrates the phase transition property exhibited by certain amino acid sequences.
  • FIG. 2 illustrates the phase transition property exhibited by certain amino acid sequences.
  • Phase transition can be observed as an increase in turbidity.
  • FIG. 3 illustrates, without wishing to be bound by theory, a potential mechanism for phase transition, driven by exclusion of a water shell and formation of hydrogen bonds at a temperature above the phase transition temperature for a given concentration.
  • FIG. 4 shows that the ELP4 series (about 120 structural units) transitions at 37° C. at a protein concentration of less than about 0.01 mg/mL, allowing for sustained release formulations of low protein concentration. At higher concentrations the sustained release will be sufficiently slow to provide a depot like formulation.
  • FIG. 5 shows that the ELP1 series transitions between 35 and 37° C. at relatively high protein concentration of about 10 mg/mL to about 100 mg/mL, or more, allowing for sustained release formulations with relatively high amounts of active agents.
  • PB1023 GLP-1, A8G,7-37, ELP1-120
  • PB1046 GLP-1, A8G,7-37, ELP4-120
  • Table 1 shows determination of phase transition for formulations of PB1023 and PB1046, varied by protein concentration and ionic strength. As shown, formulations of at least 50 mg/mL PB1023 and with an ionic strength of at least that of 10 mM His and 55 mM NaCl, transitioned at 37° C. (with an approximate transition temperature of 35.5° C.). A formulation of 25 mg/mL of PB-1023 and an ionic strength of about normal saline also transitioned at 37° C. Formulations even as low as 1 mg/mL of PB1046 and having an ionic strength similar to normal saline transitioned at 37° C.
  • Table 4 shows some buffer formulations in accordance with certain embodiments of the invention.
  • FIG. 6 shows a summary of pharmacokinetic parameters for GLP-1/ELP1-120 (also referred to herein as PB1023 or (Glymera) after SC administration of 0.3, 0.6, 0.9 and 1.35 mg/kg to adult subjects with type 2 diabetes mellitus.
  • FIG. 7 shows the mean serum concentrations of GLP-1/ELP1-120 (also referred to herein as PB1023 or Glymera) after s.c. administration on day 0 of 0.3, 0.6, 0.9 and 1.35 mg/kg to adult subjects with type 2 diabetes mellitus (semi-logarithmic axes).
  • FIG. 8 shows a type 2 diabetes mellitus: Glymera program overview pharmacokinetics crossover study. Mean serum concentrations of Glymera following s.c. administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with type 2 diabetes mellitus are shown (semi-logarithmic axes). It is noted that that the time courses for mean serum distribution for the 50 mg/mL and 100 mg/mL are nearly equivalent on the whole, except that the 100 mg/mL dose bursts into the blood stream slower than the 50 mg/mL dose (i.e. the 100 mg/mL data set has a slower rate of rise).
  • FIG. 9 shows a summary of pharmacokinetic parameters for ELP1-120 (also referred to herein as PB1023 or Glymera) after s.c. administration of 90 mg as 50 mg/mL, and 100 mg/mL formulations to adult subjects with type 2 diabetes mellitus.
  • the geometric mean ratios (GMR) for Cmax, AUC(0 ⁇ t), and AUC(inf) for the comparison of the 100 mg/mL to 50 mg/mL formulations ranged from 94.79% to 99.97% and all associated 90% confidence intervals (CI) were contained within 80.00% and 125.00%, demonstrating bioequivalence between the 2 formulations.
  • PB1023 The 100 mg/mL formulation of PB1023 was bioequivalent to the 50 mg/mL formulation after administration to adult subjects with T2DM.
  • Administration of PB1023 as a cold formulation (2° to 8° C.) resulted in a significant decrease in absorption.
  • a hypotonic formulation (containing 20 mM histidine only) of PB1023 Injection at a concentration of 50 mg/mL (formulated in 20 mM histidine) was diluted to 25 mL with 0.9% sodium chloride to render the final formulation nearly isotonic prior to administration.
  • an isotonic formulation (containing 20 mM histidine and 110 mM NaCl for tonicity) at a concentration of 100 mg/mL was manufactured for use in future clinical trials.
  • This study evaluated the impact on the rate of absorption when the 100 mg/mL formulation is injected cold (2° to 8° C.).
  • a purpose of this study was to compare the pharmacokinetic profile of PB1023 Injection after administration of the two formulations and the effect of cold administration on absorption.
  • Blood samples were collected before and 1, 4, 8, 12, 24, 48, 72, 120, 168, and 240 hours after dosing. Blood samples were centrifuged and the resultant serum was transferred to two (2) clearly labeled polypropylene tubes and stored frozen until assay.
  • AUC ⁇ ( inf ) AUC ⁇ ( 0 - t ) + C tf ⁇ ⁇ ⁇ z
  • C tf is the final concentration LOQ.
  • Clearance (CL/F) and volume of distribution (Vz/F), uncorrected for bioavailability (F) were calculated according to
  • Comparison of the kinetic parameters Cmax, AUC(04), and AUC(inf) for PB1023 between the test (100 mg/mL) and reference (50 mg/mL) formulations was done using an analysis of variance statistical model (ANOVA) with sequence, subject within sequence, treatment, and period as the classification variables, using the natural logarithms of the data.
  • Comparison of Cmax, AUC(0 ⁇ t), and AUC(inf) after administration of PB1023 as a cold formulation and at room temperature was done using an ANOVA with subject and treatment as the classification variables, using the natural logarithms of the data.
  • the within-subject coefficient of variation (WSCV) of each natural log-transformed parameter was calculated according to
  • MSE is the mean squared error from the analysis of variance.
  • the analysis population was therefore comprised of 10 subjects for the comparison of 100 mg/mL, and 50 mg/mL and 8 subjects for the comparison of 100 mg/mL, as room temperature formulation and as a cold formulation.
  • the mean serum PB1023 concentrations increased at a faster rate after administration of the 50 mg/mL formulation compared to the 100 mg/mL formulation; this was also observed for the majority of the individual subjects.
  • mean concentrations from 48 hours onward were essentially the same for both formulations ( FIG. 12 ).
  • the mean values for Cmax, AUC(0 ⁇ t), and AUC(inf) were comparable for both formulations ( FIG. 14 ) with GMRs ranging from 94.79% to 99.97% and all associated 90% CI were with 80.00% to 125.00% ( FIG. 1 ), demonstrating bioequivalence between the 2 formulations.
  • the median Tmax was similar for the 50 mg/mL and 100 mg/mL formulations (49.4 h and 48.4 h, respectively) as were the mean values for t1 ⁇ 2 and CL/F ( FIG. 14 )
  • the mean serum PB1023 concentrations were lower when the 100 mg/mL formulation was administered under cold (2° to 8° C.) compared to room temperature conditions; this was also observed for the majority but not all of the individual subjects.
  • the mean values for Cmax, AUC(0 ⁇ t), and AUC(inf) were lower when PB1023 was administered cold for both formulations ( FIG. 18 ) with GMRs of 68.66%, 79.90%, and 73.30%, respectively, and the lower limits of all 3 CIs were substantially below 80.00% ( FIG. 19 ). This demonstrates a significant decrease in absorption when PB1023 was administered cold.
  • the median Tmax was about 50% longer when PB1023 was injected as a cold formulation ( FIG. 18 ), suggesting, but not wishing to be bound by theory, a slower rate of absorption in addition to the lower bioavailability.
  • the mean t1 ⁇ 2 increased from 30.8 ⁇ 4.95 h to 40.2 ⁇ 20.1 h ( FIG. 18 ), most likely a consequence of the slower absorption and “flip-flop” kinetics.
  • PB1023 The 100 mg/mL, formulation of PB1023 was bioequivalent to the 50 mg/ml, formulation after administration to adult subjects with T2DM.
  • Administration of PB1023 as a cold formulation (2° to 8° C.) resulted in a significant decrease in absorption.
  • Transition Drug/Formulation Buffer Formulation waterbath Temperature 50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 Yes ⁇ 37° C. 20 mM Histidine 50 mM NaCl 50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 ⁇ 37° C. 20 mM Histidine 40 mM NaCl 50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 ⁇ 37° C.

Landscapes

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

Abstract

The present invention provides pharmaceutical formulations for sustained release when administered at cold temperatures, and methods for delivering a treatment regimen with a combination of sustained release and long half-life formulations. The invention provides improved pharmacokinetics for peptide and small molecule drugs.

Description

    PRIORITY
  • This application claims priority to U.S. Provisional Application No. 61/728,318, filed Nov. 20, 2012, which is hereby incorporated by reference in its entirety.
  • FIELD OF INVENTION
  • The present invention relates to pharmaceutical formulations for sustained release when administered at cold temperatures, and methods for delivering a treatment regimen with the sustained release formulations.
  • BACKGROUND
  • The effectiveness of peptide and small molecule drugs is often limited by the half-life of such drugs in the circulation, as well as difficulties in obtaining substantially constant plasma levels. For example, the incretin GLP-1 must be administered at relatively high doses to counter its short half-life in the circulation, and these high doses are associated with nausea, among other things. Murphy and Bloom, Nonpeptidic glucagon- like peptide 1 receptor agonists: A magic bullet for diabetes? PNAS 104 (3):689-690 (2007). Further, the peptide agent vasoactive intestinal peptide (VIP) exhibits a half-life, in some estimates, of less than one minute, making this agent impractical for pharmaceutical use. Domschke et al., Vasoactive intestinal peptide in man: pharmacokinetics, metabolic and circulatory effects. Gut 19:1049-1053 (1978); Henning and Sawmiller, Vasoactive intestinal peptide: cardiovascular effects, Cardiovascular Research 49:27-37 (2001). A short plasma half life for peptide drugs is often due to fast renal clearance as well as to enzymatic degradation during systemic circulation.
  • Strategies for improving the pharmacokinetics of peptide and small molecule drugs are in great demand.
  • SUMMARY OF THE INVENTION
  • The present invention provides pharmaceutical formulations for sustained release, and methods for delivering a treatment regimen with the sustained release formulations. The invention thereby provides improved pharmacokinetics for peptide and small molecule drugs.
  • In one aspect, the invention provides a sustained release pharmaceutical formulation. The formulation comprises a therapeutic agent for systemic administration, where the therapeutic agent comprises an active agent and an amino acid sequence capable of forming a reversible matrix at the body temperature of a subject. The reversible matrix is formed from hydrogen bonds (e.g., intra- and/or intermolecular hydrogen bonds) as well as from hydrophobic contributions. The formulation further comprises one or more pharmaceutically acceptable excipients and/or diluents inducing the formation of the matrix upon administration. The matrix provides for a slow absorption to the circulation from an injection site. The sustained release, or slow absorption from the injection site, is due to a slow reversal of the matrix as the concentration dissipates at the injection site. Once product moves into the circulation, the formulation confers long half-life and improved stability. Thus, a unique combination of slow absorption and long half-life is achieved leading to a desirable PK profile with a shallow peak to trough ratio and/or long Tmax. In accordance with the invention, these benefits can be provided by administering cold formulations (e.g. 2-15° C., or 2-1.0° C., or 2-5° C.) of the therapeutic agent.
  • In certain embodiments, the amino acid sequence is an Elastin-Like-Protein (ELP) sequence. The ELP sequence comprises or consists of structural peptide units or sequences that are related to, or mimics of, the elastin protein. The amino acid sequence may exhibit a visible and reversible inverse phase transition with the selected formulation. That is, the amino acid sequence may be structurally disordered and highly soluble in the formulation below a transition temperature (Tt), but exhibit a sharp (2-3° C. range) disorder-to-order phase transition when the temperature of the formulation is raised above the Tt. In addition to temperature, length of the amino acid polymer, amino acid composition, ionic strength, pH, pressure, selected solvents, presence of organic solutes, temperature, and protein concentration may also affect the transition properties, and these may be tailored for the desired absorption profile. Other exemplary sequences or structures for the amino acid sequence forming the matrix are disclosed herein.
  • In various embodiments, the active agent for systemic administration is a protein or peptide, which may have a short circulatory half-fife, such as from about 30 seconds to about 1 hour, to about 2 hours, or to about 5 hours. In some embodiments, the protein or peptide has a circulatory half-life of from 30 seconds to about 10 hours. The therapeutic agent may be a recombinant fusion protein between the protein active agent and the amino acid sequence capable of forming the matrix. Exemplary peptide active agents include GLP-1 receptor agonists (e.g., GLP-1 or derivative thereof), glucagon receptor agonists (e.g. glucagon, oxyntomodulin or derivatives thereof), VPAC2 selective agonists (e.g. vasoactive intestinal peptide (VIP) or a derivative thereof), (GIP receptor agonists (e.g. glucose-dependent insulinotropic peptide (GIP) or a derivative thereof) or insulin or a derivative thereof. Alternatively, the protein active agent is a clotting factor, such as Factor VII, Factor VIII, or Factor IX. Other protein and small molecule drugs for delivery in accordance with the invention are disclosed herein. By providing a slow absorption from the injection site, renal clearance and degradation can be controlled thereby achieving the desired PK profile.
  • In another aspect, the invention provides a method for delivering a sustained release regimen of an active agent. The method comprises administering the formulation described herein to a subject in need, wherein the formulation is administered from about 1 to about 8 times per month. In some embodiments, the formulation is administered about weekly, and may be administered subcutaneously or intramuscularly (for example). In some embodiments, the site of administration is not a pathological site, that is, the therapeutic agent is not administered directly to the intended site of action.
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the phase transition (as shown by an increase in turbidity) of an ELP1 protein, induced by a change in temperature to 37° C. or above. This property provides for a slow absorption from an injection site.
  • FIG. 2 shows the phase transition (as shown by an increase in turbidity) of an ELP4 protein, induced by a change in temperature to 25° C. or above. This property provides for a depot-like delivery.
  • FIG. 3 illustrates, without wishing to be bound by theory, a potential mechanism for the observed transition, in which a water shell is excluded under certain conditions, allowing for hydrogen bonds to form.
  • FIG. 4 shows that the ELP4 series transitions at 37° C. at a protein concentration of less than about 0.01 mg/ml, allowing for sustained release formulations of low protein concentration, for example, at the injection site
  • FIG. 5 shows that the ELP1 series transitions at just below 37° C. at relatively high protein concentration of about 10 mg/ml or more, allowing for sustained release formulations with relatively high amounts of active agents.
  • FIG. 6 shows a summary of pharmacokinetic parameters for Glp-1/ELP1-120 (also referred to herein as PB1023 or Glymera) after SC administration of 0.3, 0.6, 0.9 and 1.35 mg/kg to adult subjects with type 2 diabetes mellitus.
  • FIG. 7 shows the mean serum concentrations of Glp-1/ELP1-120 (also referred to herein as PB1023 or Glymera) after s.c. administration on day 0 of 0.3, 0.6, 0.9 and 1.35 mg/kg to adult subjects with type 2 diabetes mellitus (semi-logarithmic axes).
  • FIG. 8 shows the type 2 diabetes mellitus: Glymera program overview. pharmacokinetics crossover study. Mean serum concentrations of Glymera following s.c. administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with type 2 diabetes mellitus are shown (semi-logarithmic axes).
  • FIG. 9 shows a summary of pharmacokinetic parameters for Glymera after s.c. administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with type 2 diabetes mellitus.
  • FIG. 10 shows a summary of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as 50 mg/mL and 100 mg/mL formulations at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM.
  • FIG. 11 shows a statistical comparison of pharmacokinetic parameters for PB1023 alter SC administration of 90 mg as 50 mg/mL and 100 mg/mL formulations at room temperature and 100 mg/mL, cold (2° to 8° C.) to adult subjects with T2DM.
  • FIG. 12 shows the mean±standard deviation serum concentrations of PB1023 after SC administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with T2DM—linear axes.
  • FIG. 13 shows the mean±standard deviation serum concentrations of PB1023 after SC administration of 90 mg as 50 mg/mL, and 100 mg/mL formulations to adult subjects with T2DM—semi-logarithmic axes.
  • FIG. 14 shows a summary of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as 50 mg/mL, and 100 mg/mL, formulations to adult subjects with T2DM.
  • FIG. 15 shows a statistical comparison of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with T2DM.
  • FIG. 16 shows the mean±standard deviation serum concentrations of PB1023 after SC administration of 90 mg as the 100 mg/mL formulation at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM linear axes.
  • FIG. 17 shows the mean±standard deviation serum concentrations of PB1023 after SC administration of 90 mg as the 100 mg/mL, formulation at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM semi-logarithmic axes.
  • FIG. 18 shows a summary of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as the 100 mg/mL formulation at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM.
  • FIG. 19 shows a statistical comparison of pharmacokinetic parameters for PB1023 after SC administration of 90 mg as the 100 mg/mL formulation at room temperature and 100 mg/mL cold (2° to 8° C.) to adult subjects with T2DM.
  • FIG. 20A shows the human proinsulin sequence. The proinsulin sequence consists of the B and A chains linked with the C peptide. The C peptide is removed to form mature insulin following enzymatic cleavage at the two adjacent dibasic sites (underlined in italics).
  • FIG. 20B shows the amino acid sequence of a proinsulin ELP1-120 fusion protein. The proinsulin sequence (underlined) is fused to the ELP1-120 sequence. The amino acid sequence optionally includes an initiation methionine residue at the N terminus.
  • FIG. 21 shows the amino acid sequence of a GLP-1/ELP fusion protein as described herein.
  • DETAILED DESCRIPTION
  • The present invention provides pharmaceutical formulations for sustained release, and methods for delivering a treatment regimen with the sustained release formulations. The invention thereby provides improved pharmacokinetics for peptide and small molecule drugs, including a relatively flat PK profile with a low ratio of peak to trough, and/or a long Tmax. The PK profile can be maintained with a relatively infrequent administration schedule, such as from one to eight injections per month in some embodiments.
  • In one aspect, the invention provides a sustained release pharmaceutical formulation. The formulation comprises a therapeutic agent for systemic administration, where the therapeutic agent comprises an active agent and an amino acid sequence capable of forming a matrix at the body temperature of a subject. The reversible matrix is formed from hydrogen bonds (e.g., intra- and/or intermolecular hydrogen bonds) as well as from hydrophobic contributions. The formulation further comprises one or more pharmaceutically acceptable excipients and/or diluents inducing the formation of the matrix upon administration. The matrix provides for a slow absorption to the circulation from an injection site, and without being bound by theory, this slow absorption is due to the slow reversal of the matrix as protein concentration decreases at the injection site. The slow absorption profile provides for a flat PK profile, as well as convenient and comfortable administration regimen. For example, in various embodiments, the plasma concentration of the active agent over the course of days (e.g., from 2 to about 60 days, or from about 4 to about 30 days) does not change by more than a factor of 10, or by more than a factor of about 5, or by more than a factor of about 3. Generally, this flat PK profile is seen over a plurality of (substantially evenly spaced) administrations, such as at least 2, at least about 5, or at least about 10 administrations of the formulation. In some embodiments, the slow absorption is exhibited by a Tmax (time to maximum plasma concentration) of greater than about 5 hours, greater than about 10 hours, greater than about 20 hours, greater than about 30 hours, or greater than about 50 hours.
  • The sustained release, or slow absorption from the injection site, is controlled by the amino acid sequence capable of forming a hydrogen-bonded matrix at the body temperature of the subject, as well as the components of the formulation.
  • In some embodiments, the amino acid sequence contains structural units that farm hydrogen-bonds through protein backbone groups and/or side chain groups, and which may contribute hydrophobic interactions to matrix formation. In some embodiments, the amino acids side chains do not contain hydrogen bond donor groups, with hydrogen bonds being formed substantially through the protein backbone. Exemplary amino acids include proline, alanine, valine, glycine, and isoleucine, and similar amino acids. In some embodiments, the structural units are substantially repeating structural units, so as to create a substantially repeating structural motif, and substantially repeating hydrogen-bonding capability. In these and other embodiments, the amino acid sequence contains at least 10%, at least 20%, at least 40%, or at least 50% proline, which may be positioned in a substantially repeating pattern. In this context, a substantially repeating pattern means that at least 50% or at least 75% of the proline residues of the amino acid sequence are part of a definable structural unit. In still other embodiments, the amino acid sequence contains amino acids with hydrogen-bond donor side chains, such as serine, threonine, and/or tyrosine. In some embodiments, the repeating sequence may contain from one to about four proline residues, with remaining residues independently selected from non-polar residues, such as glycine, alanine, leucine, isoleucine, and valine. Non-polar or hydrophobic residues may contribute hydrophobic interactions to the formation of the matrix.
  • The amino acid sequences may form a “gel-like” state upon injection at a temperature higher than the storage temperature. Exemplary sequences have repeating peptide units, and/or may be relatively unstructured at the lower temperature, and achieve a hydrogen-bonded, structured, state at the higher temperature.
  • In some embodiments, the amino acid sequence capable of forming the matrix at body temperature is a peptide having repeating units of from four to ten amino acids. The repeating unit may form one, two, or three hydrogen bonds in the formation of the matrix. In certain embodiments, the amino acid sequence capable of forming the matrix at body temperature is an amino acid sequence of silk, elastin, collagen, or keratin, or mimic thereof, or an amino acid sequence disclosed in U.S. Pat. No. 6,355,776, which is hereby incorporated by reference.
  • In certain embodiments, the amino acid sequence is an Elastin-Like-Protein (ELP) sequence. The ELP sequence comprises or consists of structural peptide units or sequences that are related to, or mimics of, the elastin protein. The ELP sequence is constructed from structural units of from three to about twenty amino acids, or in some embodiments, from four to ten amino acids, such as four, five or six amino acids. The length of the individual structural units may vary or may be uniform. Exemplary structural units include units defined by SEQ ID NOS: 1-12 (below), which may be employed as repeating structural units, including tandem-repeating units, or may be employed in some combination. Thus, the ELP may comprise or consist essentially of structural unit(s) selected from SEQ ID NOS: 1-12, as defined below.
  • In some embodiments, including embodiments in which the structural units are ELP units, the amino acid sequence comprises or consists essentially of from about 10 to about 500 structural units, or in certain embodiments about 50 to about 200 structural units, or in certain embodiments from about 80 to about 200 structural units, or from about 80 to about 150 structural units, such as one or a combination of units defined by SEQ ID NOS: 1-12. Thus, the structural units collectively may have a length of from about 50 to about 2000 amino acid residues, or from about 100 to about 800 amino acid residues, or from about 200 to about 700 amino acid residues, or from about 400 to about 600 amino acid residues.
  • The amino acid sequence may exhibit a visible and reversible inverse phase transition with the selected formulation. That is, the amino acid sequence may be structurally disordered and highly soluble in the formulation below a transition temperature (Tt), but exhibit a sharp (2-3° C. range) disorder-to-order phase transition when the temperature of the formulation is raised above the Tt. In addition to temperature, length of the amino acid polymer, amino acid composition, ionic strength, pH, pressure, temperature, selected solvents, presence of organic solutes, and protein concentration may also affect the transition properties, and these may be tailored in the formulation for the desired absorption profile. Absorption profile can be easily tested by determining plasma concentration or activity of the active agent over time.
  • In certain embodiments, the ELP component(s) may be formed of structural units, including but not limited to:
      • (a) the tetrapeptide Val-Pro-Gly-Gly, or VPGG (SEQ ID NO: 1);
      • (b) the tetrapeptide Ile-Pro-Gly-Gly, or IPGG (SEQ ID NO: 2);
      • (c) the pentapeptide Val-Pro-Gly-X-Gly (SEQ ID NO: 3), or VPGXG, where X is any natural or non-natural amino acid residue, and where X optionally varies among polymeric or oligomeric repeats;
      • (d) the pentapeptide Ala-Val-Gly-Val-Pro, or AVGVP (SEQ ID NO: 4);
      • (e) the pentapeptide Ile-Pro-Gly-X-Gly, or IPDXO (SEQ ID NO: 5), where X is any natural or non-natural amino acid residue, and where X optionally varies among polymeric or oligomeric repeats;
      • (e) the pentapeptide Ile-Pro-Gly-Val-Gly, or IPGVG (SEQ ID NO: 6);
      • (f) the pentapeptide Leu-Pro-Gly-X-Gly, or LPGXG (SEQ ID NO: 7), where X is any natural or non-natural amino acid residue, and where X optionally varies among polymeric or oligomeric repeats;
      • (g) the pentapeptide Leu-Pro-Gly-Val-Gly, or LPGVG (SEQ ID NO: 8);
      • (h) the hexapeptide Val-Ala-Pro-Gly-Val-Gly, or VAPGVG (SEQ ID NO: 9):
      • (i) the octapeptide Gly-Val-Gly-Val-Pro-Gly-Val-Gly, or GVGVPGVG (SEQ ID NO: 10);
      • (j) the nonapeptide Val-Pro-Gly-Phe-Gly-Val-Gly-Ala-Gly, or VPGFGVGAG (SEQ ID NO: 11); and
      • (k) the nonapeptides Val-Pro-Gly-Val-Gly-Val-Pro-Gly-Gly, or VPGVGVPGG (SEQ ID NO: 12).
  • Such structural units defined by SEQ ID NOS:1-12 may firm structural repeat units, or may be used in combination to form an ELP. In some embodiments, the ELP component is formed entirely (or almost entirely) of one or a combination of (e.g., 2, 3 or 4) structural units selected from SEQ ID NOS: 1-12. In other embodiments, at least 75%, or at least 80%, or at least 90% of the ELP component is formed from one or a combination of structural units selected from SEQ ID NOS: 1-12, and which may be present as repeating units.
  • In certain embodiments, the ELP contains repeat units, including tandem repeating units, of Val-Pro-Gly-X-Gly (SEQ ID NO: 3), where X is as defined above, and where the percentage of Val-Pro-Gly-X-Gly (SEQ ID NO: 3) units taken with respect to the entire ELP component (which may comprise structural units other than VPGXG (SEQ ID NO: 3)) is greater than about 50%, or greater than about 75%, or greater than about 85%, or greater than about 95% of the ELP. The ELP may contain motifs of 5 to 15 structural units (e.g. about 10 structural units) of SEQ ID NO: 3, with the guest residue X varying among at least 2 or at least 3 of the units in the motif. The guest residues may be independently selected, such as from non-polar or hydrophobic residues, such as the amino acids V, I, L, A, G, and W (and may be selected so as to retain a desired inverse phase transition property).
  • In some embodiments, the ELP may form a 13-turn structure. Exemplary peptide sequences suitable tier creating a β-turn structure are described in International Patent Application PCT/US96/05186, which is hereby incorporated by reference in its entirety. For example, the fourth residue (X) in the sequence VPGXG (SEQ ID NO: 3), can be altered without eliminating the formation of a β-turn.
  • The structure of exemplary ELPs may be described using the notation ELPk [XiYj-n], where k designates a particular ELP repeat unit, the bracketed capital letters are single letter amino acid codes and their corresponding subscripts designate the relative ratio of each guest residue X in the structural units (where applicable), and n describes the total length of the [[P in number of the structural repeats. For example, ELP1 [V5A2G3-10] designates an ELP component containing 10 repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is valine, alanine, and glycine at a relative ratio of about 5:2:3; ELP1 [K1V2F1-4] designates an ELP component containing 4 repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is lysine, valine, and phenylalanine at a relative ratio of about 1:2:1; ELP1 [K1V7F1-9] designates a polypeptide containing 9 repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is lysine, valine, and phenylalanine at a relative ratio of about 1:7:1; ELP1 [V-5] designates a polypeptide containing 5 repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is valine; ELP1 [V-20] designates a polypeptide containing 20 repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is valine; ELP2 [5] designates a polypeptide containing 5 repeating units of the pentapeptide AVGVP (SEQ ID NO: 4); ELP3 [V-5] designates a polypeptide containing 5 repeating units of the pentapeptide IPGXG (SEQ ID NO: 5), where X is valine; ELP4 [V-5] designates a polypeptide containing 5 repeating units of the pentapeptide LPGXG (SEQ ID NO: 7), where X is valine.
  • With respect to ELP, the Tt is a function of the hydrophobicity of the guest residue. Thus, by varying the identity of the guest residue(s) and their mole fraction(s), ELPs can be synthesized that exhibit an inverse transition over a broad range. Thus, the Tt at a given ELP length may be decreased by incorporating a larger fraction of hydrophobic guest residues in the ELP sequence. Examples of suitable hydrophobic guest residues include valine, leucine, isoleucine, phenylalanine, tryptophan and methionine. Tyrosine, which is moderately hydrophobic, may also be used. Conversely, the It may be increased by incorporating residues, such as those selected from: glutamic acid, cysteine, lysine, aspartate, alanine, asparagine, serine, threonine, glycine, arginine, and glutamine.
  • For polypeptides having a molecular weight >100,000, the hydrophobicity scale disclosed in PCT/US96/05186 (which is hereby incorporated by reference in its entirety) provides one means far predicting the approximate Tt of a specific ELP sequence. For polypeptides having a molecular weight <100,000, the Tt may be predicted or determined by the following quadratic function: Tt=M0+M1X+M2X2 where X is the MW of the fusion protein, and M0=116.21; M1=−1.7499; M2=0.010349.
  • The ELP in some embodiments is selected or designed to provide a Tt ranging from about 10 to about 37° C. at formulation conditions, such as from about 20 to about 37° C., or from about 25 to about 37° C. In some embodiments, the transition temperature at physiological conditions (e.g., 0.9% saline) is from about 34 to 36° C., to take into account a slightly lower peripheral temperature.
  • In certain embodiments, the amino acid sequence capable of farming the hydrogen-bonded matrix at body temperature comprises [VPGXG]90, where each X is selected from V, G, and A, and wherein the ratio of V:G:A may be about 5:3:2. For example, the amino acid sequence capable of forming the hydrogen-bonded matrix at body temperature may comprise [VPGXG]120, where each X is selected from V, G, and A, and wherein the ratio of V:G:A may be about 5:3:2. As shown herein, 120 structural units of this ELP can provide a transition temperature at about 37° C. with about 5 to 15 mg/ml. (e.g., about 10 mg/ml) of protein. At concentrations of about 50 to about 100 mg/mL the phase transition temperature is about 35.5 degrees centigrade (just below body temperature), which allows for peripheral body temperature to be just less than 37° C.
  • Alternatively, the amino acid sequence capable of forming the matrix at body temperature comprises [VPGVG]90, or [VPGVG]120. As shown herein, 120 structural units of this ELP can provide a transition temperature at about 37° C. with about 0.005 to about 0.05 mg/ml (e.g., about 0.01 mg/ml) of protein.
  • Elastin-like-peptide (ELP) protein polymers and recombinant fusion proteins can be prepared as described in U.S. Patent Publication No. 2010/0022455, which is hereby incorporated by reference.
  • In other embodiments, the amino acid sequence capable of forming the matrix at body temperature may include a random coil or non-globular extended structure. For example, the amino acid sequence capable of forming the matrix at body temperature may comprise an amino acid sequence disclosed in U.S. Patent Publication No. 2008/0286808, WIPO Patent Publication No. 2008/155134, and U.S. Patent Publication No. 2011/0123487, each of which is hereby incorporated by reference.
  • For example, in some embodiments the amino acid sequence comprises an unstructured recombinant polymer of at least 40 amino acids. For example, the unstructured polymer may be defined where the sum of glycine (G), aspartate (D), alanine (A), serine (S), threonine (T), glutamate (E) and proline (P) residues contained in the unstructured polymer, constitutes more than about 80% of the total amino acids. In some embodiments, at least 50% of the amino acids are devoid of secondary structure as determined by the Chou-Fasman algorithm. The unstructured polymer may comprise more than about 100, 150, 200 or more contiguous amino acids. In some embodiments, the amino acid sequence forms a random coil domain. In particular, a polypeptide or amino acid polymer having or forming “random coil conformation” substantially lacks a defined secondary and tertiary structure.
  • In various embodiments, the intended subject is human, and the body temperature is about 37° C., and thus the therapeutic agent is designed to provide a sustained release at this temperature. A slow release into the circulation with reversal of hydrogen bonding and/or hydrophobic interactions is driven by a drop in concentration as the product diffuses at the injection site, even though body temperature remains constant. In other embodiments, the subject is a non-human mammal, and the therapeutic agent is designed to exhibit a sustained release at the body temperature of the mammal, which may be from about 30 to about 40° C. in some embodiments, such as for certain domesticated pets (e.g., dog or cat) or livestock (e.g., cow, horse, sheep, or pig). Generally, the Tt is higher than the storage conditions of the formulation. The storage conditions may be from about 2 to about 22° C., including about 2 to about 3° C., about 2 to about 4° C., about 2 to about 5° C., about 2 to about 6° C., about 2 to about 7° C., about 2 to about 8° C., about 2 to about 10° C., about 2 to about 12° C., about 2 to about 14° C., about 2 to about 16° C., about 10 to about 25° C., or from 15 to 22° C., such that the therapeutic agent remains in solution for injection and exhibits the desired pharmacokinetic behavior. The formulation may be administered at the about storage temperature.
  • The therapeutic agent is generally for “systemic delivery,” meaning that the agent is not delivered locally to a pathological site or a site of action. Instead, the agent is absorbed into the bloodstream from the injection site, where the agent acts systemically or is transported to a site of action via the circulation.
  • In various embodiments, the active agent is a protein or peptide, which may have a short circulatory half-life, such as from about 30 seconds to about 1 hour. The therapeutic agent may be a recombinant fusion protein between the protein active agent and the amino acid sequence capable of forming the hydrogen-bonded matrix at the body temperature of the subject. Exemplary peptide active agents include GIP receptor agonists such as glucose-dependent insulinotropic peptide (GIP) or a derivative thereof. Further exemplary peptide active agents include GLP1 receptor agonists such as GLP-1 or derivative thereof (including GLP1 7-36 or GLP1 7-37), or exendin or derivative thereof. GLP1 7-36 has the following amino acid sequence: HAEGTFTSDVSSYLEGQAAKEFIAWLVKGR. Exendin-4 has the following amino acid sequence: HGEGTFTSDLSKQMEEEAVRLFEWLKNGGPSSGAPPPS. In other embodiments, the protein or peptide agent is a glucagon receptor agonist (including glucagon, oxyntomodulin or a derivative thereof). Oxyntomodulin, has the amino acid sequence HSQGTFTSDYSKYLDSRRAQDFVQWLMNTKRNKNNIA. Glucagon has the amino acid sequence HSQGTFTSDYSKYLDSRRAQDFVQWLMNT. GLP-2 has the amino acid sequence HADGSFSDEMNTILDNLAARDFINWLIQTKITD. The amino acid sequence of GIP is YAEGTFISDYSIAMDKIRQQDFVNWLLAQ. In some embodiments, the GLP-1 receptor agonist is a dual agonist having an amino acid sequence described in US 2011/0257092, which is hereby incorporated by reference in its entirety. Other dual or multi receptor agonists are described in US 2011/016602 and US 2010/00190701, each of which is hereby incorporated by reference, in particular with regard to the structures and sequences of GLP-1 receptor co-agonists described therein. Additional descriptions of GLP-1 receptor co-agonists can be found in Pocai A et al., Glucagon-Like Peptide 1/Glucagon Receptor Dual Agonism Reverses Obesity in Mice, Diabetes 58:2258-2266 (2009) and Patterson J T et al., Functional association of the N-terminal residues with the central region in glucagon-related peptides, J. Pept. Sci. 17:659-666 (2011), each of which are hereby incorporated by reference in their entirety. In another embodiment, the invention provides for a co-formulation of any two of a GLP1 receptor agonist, a glucagon receptor agonist, and a GIP receptor agonist. In other embodiments, the protein or peptide agent is a VPAC2 selective agonist, such as vasoactive intestinal peptide (VIP) or a derivative thereof. Alternatively, the protein active agent is a clotting factor, such as Factor VII, Factor VIII, or Factor IX, or in other embodiments is insulin (e.g., single chain insulin or an A chain or a B chain fusion protein, as described in U.S. Provisional Application No. 61/563,985, which is hereby incorporated by reference) or a monoclonal antibody or single chain antibody. Alternatively, the active agent is as described in U.S. Patent Publication No. 2011/0123487, which is hereby incorporated by reference. Exemplary therapeutic agents in accordance with the invention include GLP-1 (A8G,7-37) ELP1-120 (referred to herein as PB1023) or GLP-1 (A8G,7-37) ELP4-120 (PB1046). By providing a slow absorption from the injection site, renal clearance and degradation can be controlled, thereby achieving the desired pK profile.
  • In various embodiments, the invention encompasses formulations which comprise a therapeutic agent for systemic administration, where the therapeutic agent comprises an active agent selected from one or more of insulin (by way of non-limiting example, single chain insulin or an A chain or a B chain fusion protein) and GLP-1 or derivative thereof (by way of non-limiting example, GLP1 7-36 or GLP1 7-37) and an amino acid sequence capable of forming a matrix at the body temperature of a subject (by way of non-limiting example, ELP). In various embodiments, the present invention encompasses a formulation, or co-formulation, of GLP-1 or derivative thereof (including GLP1 7-36 or GLP1 7-37) fused to ELP and insulin (by way of non-limiting example, single chain insulin or an A chain or a B chain fusion protein) fused to ELP. In some embodiments, these formulations are administered cold (by way of non-limiting example, 2-10° C.) or at room temperature as described herein.
  • In various embodiments, the invention encompasses doses and/or regimens such as those that do not induce substantial appetite suppression in a patient and/or those that do not induce substantial nausea in the patient, such as those described in PCT/US12/44383, which is hereby incorporated by reference.
  • In other embodiments, the therapeutic agent is a chemical conjugate between the active agent and the amino acid sequence capable of forming the matrix at the body temperature of the subject. For example, the active agent may be a chemotherapeutic agent, such as a chemotherapeutic agent selected from methotrexate, daunomycin, mitomycin, cisplatin, vincristine, epirubicin, fluorouracil, verapamil, cyclophosphamide, cytosine arabinosidc, aminopterin, bleomycin, mitomycin C, democolcine, etoposide, mithramycin, chlorambucil, melphalan, daunoubicin, doxorubicin, tamoxifen, paclitaxel, vinblastine, camptothecin, actinomycin 13, cytarabine, and combrestatin. Alternatively, the agent may be an immunogenic molecule, or an immunomodulator, or an anti-inflammatory agent, such as an agent described in U.S. Patent Publication No. 2009/0004104, which is hereby incorporated by reference in its entirety. Also, the agent may be an opioid molecule, such as, for example oxycodone, morphine, or codeine such as described in U.S. Provisional Application No. 61/597,898, which is hereby incorporated by reference. The chemical conjugate may be through a cleavable linker, for which numerous types are known in the art. See U.S. Pat. No. 6,326,996, which is hereby incorporated by reference in its entirety.
  • The formulation comprises one or more pharmaceutically acceptable excipients and/or diluents inducing the formation of the matrix upon administration. For example, such excipients include salts, and other excipients that may act to stabilize hydrogen bonding. Exemplary salts include alkaline earth metal salts such as sodium, potassium, and calcium. Counter ions include chloride and phosphate. Exemplary salts include sodium chloride, potassium chloride, magnesium chloride, calcium chloride, and potassium phosphate.
  • The protein concentration in the formulation is tailored to drive, along with the excipients, the formation of the matrix at the temperature of administration. For example, higher protein concentrations help drive the formation of the matrix, and the protein concentration needed for this purpose varies depending on the ELP series used. For example, in embodiments using an ELP1-120, or amino acid sequences with comparable transition temperatures, the protein is present in the range of about 1 mg/mL, to about 200 mg/mL, or is present in the range of about 5 mg/mL, to about 125 mg/mL. The therapeutic agent may be present in the range of about 10 mg/mL to about 50 mg/mL, or about 15 mg/mL to about 30 mg/mL. In embodiments using an ELP4-120, or amino acid sequences with comparable transition temperatures, the protein is present in the range of about 0.005 mg/mL to about 10 mg/mL, or is present in the range of about 0.01 mg/mL to about 5 mg/mL.
  • The therapeutic agent is formulated at a pH, ionic strength, and generally with excipients sufficient to drive the formation of the matrix at body temperature (e.g., 37° C., or at from 34 to 36° C. in some embodiments). The therapeutic agent is generally prepared such that it does not form the matrix at storage conditions. Storage conditions are generally less than the transition temperature of the formulation, such as less than about 32° C., or less than about 30° C., or less than about 27° C., or less than about 25° C., or less than about 20° C., or less than about 15° C. For example, the formulation may be isotonic with blood or have an ionic strength that mimics physiological conditions. For example, the formulation may have an ionic strength of at least that of 25 mM Sodium Chloride, or at least that of 30 mM Sodium chloride, or at least that of 40 mM Sodium Chloride, or at least that of 50 mM Sodium Chloride, or at least that of 75 mM Sodium Chloride, or at least that of 100 mM Sodium Chloride, or at least that of 150 mM Sodium Chloride. In certain embodiments, the formulation has an ionic strength less than that of 0.9% saline. In some embodiments, the formulation comprises two or more of calcium chloride, magnesium chloride, potassium chloride, potassium phosphate monobasic, sodium chloride, and sodium phosphate dibasic. The liquid formulation may comprise the components listed in Table 4, Table 5, or Table 6, and can be stored refrigerated or at room temperature.
  • The formulation can be packaged in the form of pre-dosed pens or syringes for administration once per week, twice per week, or from one to eight times per month, or alternatively filled in conventional vial and the like.
  • In exemplary embodiments, the invention provides a sustained release pharmaceutical formulation that comprises a therapeutic agent, the therapeutic agent (e.g., a peptide or protein therapeutic agent) comprising an active agent and an amino acid sequence comprising [VPGXG]90, or [VPGXG]120, where each X is selected from V, G, and A. V, G, and A may be present at a ratio of about 5:3:2. Alternatively, the amino acid sequence comprises [VPGXG]90, or [VPGXG]120. The formulation further comprises one or more pharmaceutically acceptable excipients and/or diluents for formation of a reversible matrix from an aqueous form upon administration to a human subject. The active agent in certain embodiments is GLP-1 or derivative thereof (e.g., GLP-1, A8G, 7-37), or vasoactive intestinal peptide (VIP) or a derivative thereof (e.g., having an N-terminal moiety such as a Methionine), or oxyntomodulin of a derivative thereof, or insulin or a derivative thereof. GLP-1 and derivatives thereof are disclosed in U.S. Patent Publication No. 2011/0123487, which is hereby incorporated by reference. VIP and derivatives thereof are disclosed in U.S. Patent Publication No. 2011/0178017, which is hereby incorporated by reference. Insulin and derivatives thereof are described in U.S. Provisional Application No. 61/563,985, which is hereby incorporated by reference.
  • In these embodiments, the therapeutic agent may be present in the range of about 0.5 mg/mL to about 200 mg/mL, or is present in the range of about 5 mg/mL to about 125 mg/ML. The therapeutic agent is present in the range of about 10 mg/mL to about 50 mg/nit, or the range of about 15 mg/mL, to about 30 mg/mL. The formulation may have an ionic strength of at least that of 25 mM Sodium Chloride, or at least that of 30 mg/mL sodium Chloride, or at least that of 40 mM Sodium Chloride, or at that least that of 50 mM Sodium Chloride, or at least that of 75 mM Sodium Chloride, or at least that of 100 mM Sodium Chloride. The formulation may have an ionic strength less than that of about 0.9% saline. The formulation comprises two or more of calcium chloride, magnesium chloride, potassium chloride, potassium phosphate monobasic, sodium chloride, and sodium phosphate dibasic. The formulation may comprise the components listed in Table 4, Table 5, or Table 6.
  • Other formulation components for achieving the desired stability, for example, may also be employed. Such components include one or more amino acids or sugar alcohol (e.g., mannitol), preservatives, and buffering agents, and such ingredients are well known in the art.
  • In another aspect, the invention provides a method for delivering a sustained release regimen of an active agent. The method comprises administering the formulation described herein to a subject in need, wherein the formulation is administered from about 1 to about 8 times per month (e.g., about weekly). For example, the active agent may be GLP-1 or an analog thereof, and is administered in a method described in U.S. patent application Ser. No. 13/534,836, which is hereby incorporated by reference. For example, the therapeutic agent may be GLP-1 7-36 or 7-37, alternatively having Gly at position 8, fused to ELP1 (e.g., having from about 90 to about 150 ELP units). The GLP-1 fusion may be used for the treatment of diabetes (type 1 or 2), metabolic disease, or obesity, for example, by administering to a patient in need. Alternatively, the active agent is VIP or an analog thereof, and is administered in a method described in U.S. Patent Publication No. 2011/0178017, which is hereby incorporated by reference. The VIP may have an additional moiety such as Methionine at the N-terminus to alter the receptor binding profile, as also described in U.S. Patent Publication No. 2011/0178017, which description is hereby incorporated by reference. The VIP may be fused to ELP1 (having from about 90 to about 150 ELP units). The VIP active agent finds use in a method of treating a condition selected from uncontrolled or resistant hypertension, or pulmonary arterial hypertension (PAH), and chronic obstructive pulmonary disease (COPD), among others. In other embodiments, the active agent is insulin, which finds use in methods (using the ELP-based regimens described herein) of treating diabetes, including type 1 or type 2 diabetes.
  • In some embodiments, the formulation is administered about weekly, and may be administered subcutaneously or intramuscularly. In some embodiments, the site of administration is not a pathological site, for example, is not the intended site of action.
  • In various embodiments, the plasma concentration of the active agent does not change by more than a factor of 10, or a factor of about 5, or a factor of about 3 over the course of a plurality of administrations, such as at least 2, at least about 5, or at least about 10 administrations of the formulation. The administrations are substantially evenly spaced, such as, for example, about daily, or about once per week, or from one to about five times per month.
  • In certain embodiments, the subject is a human, but in other embodiments may be a non-human mammal, such as a domesticated pet (e.g., dog or cat), or livestock or farm animal (e.g., horse, cow, sheep, or pig).
  • EXAMPLES
  • The phase transition property exhibited by certain amino acid sequences is illustrated in FIG. 1 (for ELP1) and FIG. 2 (for ELP4). Phase transition can be observed as an increase in turbidity. FIG. 3 illustrates, without wishing to be bound by theory, a potential mechanism for phase transition, driven by exclusion of a water shell and formation of hydrogen bonds at a temperature above the phase transition temperature for a given concentration.
  • FIG. 4 shows that the ELP4 series (about 120 structural units) transitions at 37° C. at a protein concentration of less than about 0.01 mg/mL, allowing for sustained release formulations of low protein concentration. At higher concentrations the sustained release will be sufficiently slow to provide a depot like formulation. FIG. 5 shows that the ELP1 series transitions between 35 and 37° C. at relatively high protein concentration of about 10 mg/mL to about 100 mg/mL, or more, allowing for sustained release formulations with relatively high amounts of active agents.
  • Various formulations were prepared for PB1023 (GLP-1, A8G,7-37, ELP1-120) and PB1046 (GLP-1, A8G,7-37, ELP4-120), at varying protein concentrations and ionic strength. Transition induced by 37° C. water bath was tested.
  • Table 1 shows determination of phase transition for formulations of PB1023 and PB1046, varied by protein concentration and ionic strength. As shown, formulations of at least 50 mg/mL PB1023 and with an ionic strength of at least that of 10 mM His and 55 mM NaCl, transitioned at 37° C. (with an approximate transition temperature of 35.5° C.). A formulation of 25 mg/mL of PB-1023 and an ionic strength of about normal saline also transitioned at 37° C. Formulations even as low as 1 mg/mL of PB1046 and having an ionic strength similar to normal saline transitioned at 37° C.
  • As shown in Table 2, Formulations of 25 mg/mL PB1023 in either: normal saline, DPBS, or 1× phosphate buffered saline, were sufficient to generate the desired transition property. Water alone did not support the desired transition property.
  • As shown in Table 3, a formulation of 25 mg/ml PB1023 transitions at 37° C. with an ionic strength equal to 50 mM NaCl.
  • Table 4, Table 5, and Table 6 show some buffer formulations in accordance with certain embodiments of the invention.
  • FIG. 6 shows a summary of pharmacokinetic parameters for GLP-1/ELP1-120 (also referred to herein as PB1023 or (Glymera) after SC administration of 0.3, 0.6, 0.9 and 1.35 mg/kg to adult subjects with type 2 diabetes mellitus.
  • FIG. 7 shows the mean serum concentrations of GLP-1/ELP1-120 (also referred to herein as PB1023 or Glymera) after s.c. administration on day 0 of 0.3, 0.6, 0.9 and 1.35 mg/kg to adult subjects with type 2 diabetes mellitus (semi-logarithmic axes).
  • FIG. 8 shows a type 2 diabetes mellitus: Glymera program overview pharmacokinetics crossover study. Mean serum concentrations of Glymera following s.c. administration of 90 mg as 50 mg/mL and 100 mg/mL formulations to adult subjects with type 2 diabetes mellitus are shown (semi-logarithmic axes). It is noted that that the time courses for mean serum distribution for the 50 mg/mL and 100 mg/mL are nearly equivalent on the whole, except that the 100 mg/mL dose bursts into the blood stream slower than the 50 mg/mL dose (i.e. the 100 mg/mL data set has a slower rate of rise).
  • FIG. 9 shows a summary of pharmacokinetic parameters for ELP1-120 (also referred to herein as PB1023 or Glymera) after s.c. administration of 90 mg as 50 mg/mL, and 100 mg/mL formulations to adult subjects with type 2 diabetes mellitus.
  • Cold Administration
  • An open label, single dose, 2-period, 2-treatment, 2-sequence crossover design study to compare the pharmacokinetic profile of PB1023 after a single dose administered by subcutaneous (SC) injection of 50 mg/mL and 100 mg/mL formulations, the latter at room temperature and cold (2° to 8° C.) undertaken. Adult subjects with Type 2 Diabetes Mellitus (T2DM) were given a single 90 mg dose of each product at room temperature according to a randomized, 2-period, 2-sequence design. Subjects then returned for a 3rd treatment, 100 mg/mL administered cold (2° to 8° C.). Ten (10) subjects were enrolled and all subjects completed the 2 randomized treatments. Eight (8) subjects returned for the 3rd period. The analysis population was therefore comprised of 10 subjects for the comparison of 100 mg/mL and 50 mg/mL and 8 subjects for the comparison of 100 mg/mL at room temperature and as a cold formulation.
  • The pharmacokinetic parameters for PB1023 and the associated statistical analyses are summarized in the FIGS. 10 and 11.
  • The geometric mean ratios (GMR) for Cmax, AUC(0−t), and AUC(inf) for the comparison of the 100 mg/mL to 50 mg/mL formulations ranged from 94.79% to 99.97% and all associated 90% confidence intervals (CI) were contained within 80.00% and 125.00%, demonstrating bioequivalence between the 2 formulations.
  • When administered cold (2° to 8° C.), there was a decrease in Cmax, AUC(0-t), and AUC(inf) with GNIRs of 68.66%, 79.90%, and 73.30%, respectively, and the lower limits of all 3 CIs were substantially below 80.00%. This demonstrates a significant decrease in absorption when PB1023 was administered cold.
  • The 100 mg/mL formulation of PB1023 was bioequivalent to the 50 mg/mL formulation after administration to adult subjects with T2DM. Administration of PB1023 as a cold formulation (2° to 8° C.) resulted in a significant decrease in absorption.
  • A hypotonic formulation (containing 20 mM histidine only) of PB1023 Injection at a concentration of 50 mg/mL (formulated in 20 mM histidine) was diluted to 25 mL with 0.9% sodium chloride to render the final formulation nearly isotonic prior to administration. To eliminate the need for tonicity adjustment by study personnel, an isotonic formulation (containing 20 mM histidine and 110 mM NaCl for tonicity) at a concentration of 100 mg/mL was manufactured for use in future clinical trials.
  • This study evaluated the impact on the rate of absorption when the 100 mg/mL formulation is injected cold (2° to 8° C.). A purpose of this study was to compare the pharmacokinetic profile of PB1023 Injection after administration of the two formulations and the effect of cold administration on absorption.
  • This was an open label, single dose, 3-period, 3-treatment, 2-sequence crossover/sequential design study of the pharmacokinetics of PB1023 after a single dose administered by subcutaneous (SC) injection of 50 mg/mL and 100 mg/mL formulations at room temperature and 100 mg/mL as a cold formulation. Adult subjects with T2DM received, according to a randomization schedule, a dose of one of the PB1023 formulations at each visit.
  • Blood samples were collected before and 1, 4, 8, 12, 24, 48, 72, 120, 168, and 240 hours after dosing. Blood samples were centrifuged and the resultant serum was transferred to two (2) clearly labeled polypropylene tubes and stored frozen until assay.
  • All pharmacokinetic parameters were calculated using non-compartmental analysis. Only those serum concentrations equal to or greater than the LOQ (9.76 mg/mL) were used in the analysis. Actual sampling times were used in all pharmacokinetic analyses. Per protocol times were used to calculate mean serum concentrations for graphical displays.
  • The maximum serum concentration (Cmax) and time to Cmax (Tmax) were taken directly from the data. The elimination rate constant, λz, was calculated as the negative of the slope of the terminal log-linear segment of the serum concentration-time curve. The range of data used for each subject and treatment was determined by visual inspection of a semi-logarithmic plot of concentration vs. time. Elimination half-life (t½) was calculated according to the following equation.
  • t 1 /2 = 0.693 λ z
  • Area under the curve from zero to the final sample with a concentration ≧LOQ [AUC(0−t)] was calculated using the linear trapezoidal method and extrapolated to infinity [AUC(inf)] using
  • AUC ( inf ) = AUC ( 0 - t ) + C tf λ z
  • where Ctf is the final concentration LOQ.
    Clearance (CL/F) and volume of distribution (Vz/F), uncorrected for bioavailability (F) were calculated according to
  • CL / F = Dose AUC ( inf ) and Vz / F = Dose AUC ( inf ) × λ z .
  • respectively.
  • All pharmacokinetic calculations were done and individual subject plasma concentration-time graphs were prepared using SAS® for Windows® Version 9.3. Graphs of mean plasma concentration vs. time were prepared using SigmaPlot for Windows Version 12.2.
  • Comparison of the kinetic parameters Cmax, AUC(04), and AUC(inf) for PB1023 between the test (100 mg/mL) and reference (50 mg/mL) formulations was done using an analysis of variance statistical model (ANOVA) with sequence, subject within sequence, treatment, and period as the classification variables, using the natural logarithms of the data. Comparison of Cmax, AUC(0−t), and AUC(inf) after administration of PB1023 as a cold formulation and at room temperature was done using an ANOVA with subject and treatment as the classification variables, using the natural logarithms of the data.
  • For both analyses, confidence intervals (CI) (90%) were constructed for the geometric mean ratio (GMR), test-to-reference, of the three parameters using the log-transformed data and the two one-sided t-tests procedure. The GMRs and CI limits were exponentiated back to the original scale.
  • The within-subject coefficient of variation (WSCV) of each natural log-transformed parameter was calculated according to

  • WSCV=100%×√{square root over (e MSE−1)}
  • where MSE is the mean squared error from the analysis of variance.
  • All statistical analyses were done using SAS® for Windows® Version 9.3.
  • Ten (10) subjects were enrolled and all subjects completed the 2 randomized treatments. Eight (8) subjects returned for the 3rd period. The analysis population was therefore comprised of 10 subjects for the comparison of 100 mg/mL, and 50 mg/mL and 8 subjects for the comparison of 100 mg/mL, as room temperature formulation and as a cold formulation.
  • Pharmacokinetics
  • Comparison of 100 mg/mL and 50 mg/mL Formulations
  • As shown in FIG. 12 (linear axes) and FIG. 13 (semi-logarithmic axes), the mean serum PB1023 concentrations increased at a faster rate after administration of the 50 mg/mL formulation compared to the 100 mg/mL formulation; this was also observed for the majority of the individual subjects. However, mean concentrations from 48 hours onward were essentially the same for both formulations (FIG. 12). The mean values for Cmax, AUC(0−t), and AUC(inf) were comparable for both formulations (FIG. 14) with GMRs ranging from 94.79% to 99.97% and all associated 90% CI were with 80.00% to 125.00% (FIG. 1), demonstrating bioequivalence between the 2 formulations.
  • The median Tmax was similar for the 50 mg/mL and 100 mg/mL formulations (49.4 h and 48.4 h, respectively) as were the mean values for t½ and CL/F (FIG. 14)
  • Comparison of the 100 mg/mL Formulation Administered Cold (2° to 8° and at Room Temperature
  • As shown FIG. 16 (linear axes) and FIG. 17 (semi-logarithmic axes), the mean serum PB1023 concentrations were lower when the 100 mg/mL formulation was administered under cold (2° to 8° C.) compared to room temperature conditions; this was also observed for the majority but not all of the individual subjects. The mean values for Cmax, AUC(0−t), and AUC(inf) were lower when PB1023 was administered cold for both formulations (FIG. 18) with GMRs of 68.66%, 79.90%, and 73.30%, respectively, and the lower limits of all 3 CIs were substantially below 80.00% (FIG. 19). This demonstrates a significant decrease in absorption when PB1023 was administered cold.
  • The median Tmax was about 50% longer when PB1023 was injected as a cold formulation (FIG. 18), suggesting, but not wishing to be bound by theory, a slower rate of absorption in addition to the lower bioavailability. The mean t½ increased from 30.8±4.95 h to 40.2±20.1 h (FIG. 18), most likely a consequence of the slower absorption and “flip-flop” kinetics.
  • The 100 mg/mL, formulation of PB1023 was bioequivalent to the 50 mg/ml, formulation after administration to adult subjects with T2DM. Administration of PB1023 as a cold formulation (2° to 8° C.) resulted in a significant decrease in absorption.
  • TABLE 1
    Initial Transition Experiments Using a 37° C. Waterbath and Visual Interpretation of Results
    Transition Cary
    Dilution Final Concentration/ in 37° C. Transition
    Drug/Formulation Buffer Formulation waterbath Temperature
    100 mg/mL PB1023 NA 100 mg/mL PB1023 Yes ~34.9° C.
    20 mM His, 110 mM NaCl 20 mM His, 110 mM NaCl
    100 mg/mL PB1023 Water 90 mg/mL Yes
    20 mM His, 110 mM NaCl 18 mM His, 99 mM NaCl
    100 mg/mL PB1023 Water 80 mg/mL Yes
    20 mM His, 110 mM NaCl 16 mM His, 88 mM NaCl
    100 mg/mL PB1023 Water 50 mg/mL Yes
    20 mM His, 110 mM NaCl 10 mM His, 55 mM NaCl
    50 mg/mL PB1023 NA 50 mg/mL PB1023 No ~49° C.
    20 mM His, 110 mM NaCl 20 mM Histidine
    50 mg/mL PB1023 Normal Saline 25 mg/mL Yes
    20 mM Histidine (0.9% NaCl) 10 mM Histidine,
    75 mM NaCl
    40 mg/mL PB1046 NA 40 mg/mL PB1046 Yes
    20 mM His, 75 mM NaCl 20 mM His, 75 mM NaCl
    40 mg/mL PB1046 Normal Saline 12 mg/mL PB1046 Yes
    20 mM His, 75 mM NaCl (0.9% NaCl)
    40 mg/mL PB1046 Normal Saline 1 mg/mL PB1046 Yes
    20 mM His, 75 mM NaCl (0.9% NaCl)
  • TABLE 2
    Transition Temperature Experiments
    Using Various Dilution Buffers
    Final Transition Cary
    Dilution Concen- in 37° C. Transition
    Drug/Formulation Buffer tration waterbath Temperature
    50 mg/mL PB1023 Water 25 mg/mL No ~51.1
    20 mM Histidine PB1023
    50 mg/mL PB1023 Normal 25 mg/mL Yes ~36.5
    20 mM Histidine Saline PB1023
    (0.9% NaCl)
    50 mg/mL PB1023 DPBS w/MG 25 mg/mL Yes
    20 mM Histidine and Ca PB1023
    50 mg/mL PB1023 DPBS w/out 25 mg/mL Yes
    20 mM Histidine MG and Ca PB1023
    50 mg/mL PB1023 IX PBS 25 mg/mL Yes
    20 mM Histidine PB1023
  • TABLE 3
    Transition Experiments Varying Salt Concentration
    Transition Cary
    Dilution Final Concentration/ in 37° C. Transition
    Drug/Formulation Buffer Formulation waterbath Temperature
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 Yes ~37° C.
    20 mM Histidine 50 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 ~37° C.
    20 mM Histidine 40 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 ~37° C.
    20 mM Histidine 30 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 Not Visible ~37° C.
    20 mM Histidine 25 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 Not Visible
    20 mM Histidine 12.5 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 ~37° C.
    20 mM Histidine 10 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 Not Visible
    20 mM Histidine 6.25 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 Not Visible
    20 mM Histidine 3.125 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 Not Visible
    20 mM Histidine 1.56 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 ~40.3° C.
    20 mM Histidine 1 mM NaCl
    50 mg/mL PB1023 NaCl and Water 25 mg/mL PB1023 Not Visible
    20 mM Histidine 0.78 mM NaCl
  • TABLE 4
    Buffer Formulation-DPBS with Mg and Ca
    Molecular Concentration
    COMPONENTS Weight (mg/L) mM
    Inorganic Salts
    Calcium Chloride 111 100 0.901
    (CaCl2) (anhyd.)
    Magnesium Chloride 203 100 0.493
    (MgCl2—6H20)
    Potassium Chloride (KCl) 75 200 2.67
    Potassium Phosphate 136 200 1.47
    monobasic (KH2PO4)
    Sodium Chloride (NaCl) 58 8000 137.93
    Sodium Phosphate dibasic 268 2160 8.06
    (Na2HPO4—7H2O)
  • TABLE 5
    Buffer Formulation-DPBS without Mg and Ca
    Molecular Concentration
    COMPONENTS Weight (mg/L) mM
    Inorganic Salts
    Potassium Chloride (KCl) 75 200 2.67
    Potassium Phosphate 136 200 1.47
    monobasic (KH2PO4)
    Sodium Chloride (NaCl) 58 8000 137.93
    Sodium Phosphate dibasic 268 2160 8.06
    (Na2HPO4—7H2O)
  • TABLE 6
    Buffer Formulation-1x PBS pH 7.4
    Molecular Concentration
    COMPONENTS Weight (mg/L) mM
    Inorganic Salts
    Potassium Phosphate 136 144 1.06
    monobasic (Kh2PO4)
    Sodium Chloride (NaCl) 58 9000 155.17
    Sodium Phosphate dibasic 268 795 2.97
    (Na2HPO4—7H2O)

Claims (50)

That is claimed is:
1. A method for delivering a sustained release regimen of an active agent to a subject in need, comprising, administering a therapeutic agent for systemic administration, the therapeutic agent comprising an active agent and an amino acid sequence capable of forming a reversible matrix at the body temperature of a subject when administered as a cold formulation, the reversible matrix formed of hydrogen bonds and/or hydrophobic interactions, and one or more pharmaceutically acceptable excipients and/or diluents inducing the formation of the matrix upon administration.
2. The method of claim 1, wherein the cold formulation is from about 2 to about 3° C., about 2 to about 4° C., about 2 to about 5° C., about 2 to about 6° C., about 2 to about 7° C., about 2 to about 8° C., about 2 to about 10° C., about 2 to about 12° C., about 2 to about 14° C., about 2 to about 15° C., about 2 to about 16° C., about 2 to about 20° C., about 10 to about 25° C., or from 15 to 22° C.
3. The method of claim 2, wherein the formulation provides a flat PK profile upon administration, as compared to the PK profile for the active agent in the absence of the amino acid sequence forming a reversible matrix.
4. The method of claim 3, wherein the PK profile has a shallow Cmax and/or low ratio of peak to trough and/or long Tmax.
5. The method of any one of claims 1 to 4, wherein the formation of the matrix reverses as protein concentration decreases.
6. The method of any one of claims 1 to 5, wherein the amino acid sequence capable of forming the matrix at or around body temperature is a repeating peptide sequence having repeating units of from four to ten amino acids.
7. The method of claim 6, wherein the repeating unit forms one, two, or three hydrogen bonds in the formation of the matrix.
8. The method of claim 6, wherein the amino acid sequence capable of forming the matrix at body temperature is an amino acid sequence of silk, elastin, collagen, or keratin, or mimic thereof.
9. The method of any one of claims 1 to 6, wherein the amino acid sequence capable of forming the matrix at body temperature comprises [VPGXG]90, where each X is selected from V, G, and A, and wherein the ratio of V:G:A may be about 5:3:2.
10. The method of claim 9, wherein the amino acid sequence capable of forming the matrix at body temperature comprises [VPGXG]120, where each X is selected from V, G, and A, and wherein the ratio of V:G:A may be about 5:3:2.
11. The method of any one of claims 1 to 6, wherein the amino acid sequence capable of forming the matrix at body temperature comprises [VPGVG]90.
12. The method of any one of claims 1 to 6, wherein the amino acid sequence capable of forming the matrix at body temperature is an elastin-like-peptide (ELP) sequence.
13. The method of any one of claims 1 to 5, wherein the amino acid sequence capable of forming the matrix at body temperature forms a random coil or non-globular extended structure.
14. The method of any one of claims 1 to 5, wherein the amino acid sequence capable of forming the matrix at body temperature is includes a random coil, or a non-globular extended structure.
15. The method of any one of claims 1 to 14, wherein the subject is human.
16. The method of any one of claims 1 to 14, wherein the subject is a non-human mammal.
17. The method of any one of claims 1 to 16, wherein the active agent is a protein.
18. The method of claim 17, wherein the therapeutic agent is a recombinant fusion protein between the protein active agent and the amino acid sequence capable of forming the matrix at the body temperature of the subject.
19. The method of claim 18, wherein the protein active agent has a circulatory half-life in the range of from about 30 seconds to about 10 hour, or about 30 seconds to about 1 hour.
20. The method of claim 19, wherein the active agent is a GLP-1 receptor agonist or derivative thereof, a VPAC2 selective agonist or a derivative thereof, a GIP receptor agonist or a derivative thereof or a glucagon receptor agonist or a derivative thereof.
21. The method of claim 19, wherein the formulation is a co-formulation comprising at least two of a GLP1 receptor agonist, a glucagon receptor agonist, and a GIP receptor agonist
22. The method of claim 19, wherein the protein active agent is a clotting factor, where the clotting factor may be Factor VII, Factor VIII, or Factor IX.
23. The method of any one of claims 1 to 22, wherein the active agent s selected from GLP-1 (A8G,7-37) ELP1-120 and GLP-1 (A8G,7-37) ELP4-120 (PB1046).
24. The method of claim 19, wherein the protein active agent is insulin or a derivative thereof.
25. The method of any one of claims 1 to 17, wherein the therapeutic agent is a chemical conjugate between the active agent and the amino acid sequence capable of forming the matrix at the body temperature of the subject.
26. The method of claim 25, wherein the active agent is a chemotherapeutic agent, such as a chemotherapeutic agent selected from methotrexate, daunomycin, mitomycin, cisplatin, vincristine, epirubicin, fluorouracil, verapamil, cyclophosphamide, cytosine arabinoside, aminopterin, bleomycin, mitomycin C, democolcine, etoposide, mithramycin, chlorambucil, melphalan, daunorubicin, doxorubicin, tamoxifen, paclitaxel, vinblastine, camptothecin, actinomycin D, cytarabine, and combrestatin.
27. The method of claim 25, wherein the agent is an immunogenic molecule, or is an immunomodulator, or is an anti-inflammatory agent.
28. The method of any one of claims 1 to 27, wherein the therapeutic agent is present in the range of about 0.5 mg/mL to about 200 mg/mL.
29. The method of claim 28, wherein the therapeutic agent is present in the range of about 5 mg/mL, to about 125 mg/mL.
30. The method of claim 28, wherein the therapeutic agent is present in the range of about 10 mg/mL to about 50 mg/mL or about 15 mg/mL to about 30 mg/mL.
31. The method of any one of claims 1 to 30, wherein the therapeutic agent does not form the phase-transitioned matrix at storage conditions.
32. The method of claim 31, wherein the storage conditions are less than about 40° C., or less than about 37° C., or less than about 30° C., or less than about 27° C., or less than about 25° C., or less than about 20° C., or less than about 15° C., or less than about 10° C., or less than about 5° C.
33. The method of any one of claims 1 to 32, wherein the formulation has an ionic strength of at least that of 25 mM Sodium Chloride, or at least that of 30 mM Sodium chloride, or at least that of 40 mM Sodium Chloride, or at least that of 50 mM Sodium Chloride, or at least that of 75 mM Sodium Chloride, or at least that of 100 mM Sodium Chloride, or at least that of 150 mM Sodium Chloride.
34. The method of any one of claims 1 to 33, wherein the formulation has an ionic strength of about 0.9% saline or less.
35. The method of claim 34, wherein the formulation comprises two or more of calcium chloride, magnesium chloride, potassium chloride, potassium phosphate monobasic, sodium chloride, and sodium phosphate dibasic.
36. The method of claim 35, wherein the formulation comprises the components listed in Table 4, Table 5, or Table 6.
37. The method of any one of claims 1 to 36, wherein the formulation is packaged in the form of pre-dosed pens or syringes for administration once per week, twice per week, or from one to five times per month.
38. The method of any one of claims 1 to 37, wherein the formulation is packaged in the form of pre-dosed pens or syringes for administration once per week, twice per week, or from one to five times per month.
39. The method of any one of claims 1 to 38, wherein the formulation is administered from about 1 to about 8 times per month.
40. The method of any one of claims 1 to 39, wherein the active agent is GLP-1 or an analog thereof, and is administered about 1 to about 8 times per month.
41. The method of any one of claims 1 to 40, wherein the active agent is VIP or an analog thereof, and is administered about 1 to about 8 times per month.
42. The method of any one of claims 39 to 41, wherein the formulation is administered about weekly.
43. The method of any one of claims 39 to 41, wherein the formulation is administered subcutaneously or intramuscularly.
44. The method of any one of claims 39 to 41, wherein the site of administration is not a pathological site.
45. A sustained release pharmaceutical formulation comprising:
a therapeutic agent, the therapeutic agent comprising an active agent and an amino acid sequence comprising [VPGXG]90, where each X is selected from V, G, and A, and one or more pharmaceutically acceptable excipients and/or diluents for formation of a reversible matrix from an aqueous form upon administration to a human subject as a cold formulation.
46. The sustained release pharmaceutical formulation of claim 45, wherein the cold formulation is from about 2 to about 3° C., about 2 to about 4° C., about 2 to about 5° C., about 2 to about 6° C., about 2 to about 7° C., about 2 to about 8° C., about 2 to about 10° C., about 2 to about 12° C., about 2 to about 14° C., about 2 to about 15° C., about 2 to about 16° C., about 2 to about 20° C., about 10 to about 25° C., or from 15 to 22° C.
47. A method for delivering a sustained release regimen of insulin to a subject in need, comprising: administering, about once weekly, a cold formulation of an insulin polypeptide fusion with an amino acid sequence capable of forming a reversible matrix at the body temperature of a subject, the reversible matrix formed of hydrogen bonds and/or hydrophobic interactions, and the formulation comprising a once weekly effective amount of the insulin fusion and one or more pharmaceutically acceptable excipients and/or diluents inducing the formation of the matrix upon administration to the subject.
48. The method of claim 47, wherein the cold formulation is administered at from about 2 to about 22° C.
49. The method of claim 47, wherein the cold formulation is administered at from about 2 to about 10° C.
50. The method of any one of claims 47 to 49, wherein the amino acid sequence capable of forming a reversible matrix at the body temperature of a subject is [VPGXG]120, where each X is selected from V, G, and A, and wherein the ratio of V:G:A is about 5:3:2.
US14/646,273 2012-11-20 2013-11-20 Formulations of active agents for sustained release Abandoned US20150290328A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/646,273 US20150290328A1 (en) 2012-11-20 2013-11-20 Formulations of active agents for sustained release

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261728318P 2012-11-20 2012-11-20
US14/646,273 US20150290328A1 (en) 2012-11-20 2013-11-20 Formulations of active agents for sustained release
PCT/US2013/071038 WO2014081849A1 (en) 2012-11-20 2013-11-20 Formulations of active agents for sustained release

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/071038 A-371-Of-International WO2014081849A1 (en) 2012-11-20 2013-11-20 Formulations of active agents for sustained release

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/355,386 Continuation US9919032B2 (en) 2012-11-20 2016-11-18 Method for administering a sustained release formulation

Publications (1)

Publication Number Publication Date
US20150290328A1 true US20150290328A1 (en) 2015-10-15

Family

ID=50776530

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/646,273 Abandoned US20150290328A1 (en) 2012-11-20 2013-11-20 Formulations of active agents for sustained release
US15/355,386 Active US9919032B2 (en) 2012-11-20 2016-11-18 Method for administering a sustained release formulation

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/355,386 Active US9919032B2 (en) 2012-11-20 2016-11-18 Method for administering a sustained release formulation

Country Status (2)

Country Link
US (2) US20150290328A1 (en)
WO (1) WO2014081849A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017192449A1 (en) * 2016-05-06 2017-11-09 Phasebio Pharmaceuticals, Inc. Elp fusion proteins for controlled and sustained release
US9919032B2 (en) 2012-11-20 2018-03-20 Phasebio Pharmaceuticals, Inc. Method for administering a sustained release formulation

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012019168A2 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
DE19177059T1 (en) 2010-10-01 2021-10-07 Modernatx, Inc. RIBONUCLEIC ACID CONTAINING N1-METHYL-PSEUDOURACILE AND USES
DE12722942T1 (en) 2011-03-31 2021-09-30 Modernatx, Inc. RELEASE AND FORMULATION OF MANIPULATED NUCLEIC ACIDS
CA2873553C (en) 2011-06-06 2020-01-28 Phasebio Pharmaceuticals, Inc. Use of modified vasoactive intestinal peptides in the treatment of hypertension
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
JP6113737B2 (en) 2011-10-03 2017-04-12 モデルナティエックス インコーポレイテッドModernaTX,Inc. Modified nucleosides, nucleotides and nucleic acids and methods for their use
MX2014007233A (en) 2011-12-16 2015-02-04 Moderna Therapeutics Inc Modified nucleoside, nucleotide, and nucleic acid compositions.
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
EP2834259A4 (en) 2012-04-02 2016-08-24 Moderna Therapeutics Inc Modified polynucleotides
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
CA2892529C (en) 2012-11-26 2023-04-25 Moderna Therapeutics, Inc. Terminally modified rna
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
CA2926218A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
ES2818824T3 (en) 2014-05-08 2021-04-14 Phasebio Pharmaceuticals Inc Compositions comprising a VIP-ELP fusion protein for use in the treatment of cystic fibrosis
US10722590B2 (en) 2014-11-21 2020-07-28 Phasebio Pharmaceuticals, Inc. ELP fusion proteins for controlled and sustained release
ES2822598T3 (en) 2015-02-09 2021-05-04 Phasebio Pharmaceuticals Inc Methods and Compositions for Treating Muscle Diseases and Disorders
WO2018232357A1 (en) 2017-06-15 2018-12-20 Modernatx, Inc. Rna formulations
EP3675817A1 (en) 2017-08-31 2020-07-08 Modernatx, Inc. Methods of making lipid nanoparticles

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080312157A1 (en) * 2005-02-11 2008-12-18 Amylin Pharmaceuticals, Inc. Gip analog and hybrid polypeptides with selectable properties

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6018030A (en) 1986-11-04 2000-01-25 Protein Polymer Technologies, Inc. Peptides comprising repetitive units of amino acids and DNA sequences encoding the same
DE69105323T2 (en) 1990-03-27 1995-06-01 Bioelastics Res Ltd Bioelastomeric drug delivery system.
US5854387A (en) 1995-04-14 1998-12-29 Bioelastics Research, Ltd. Simple method for the purification of a bioelastic polymer
US7846445B2 (en) 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
WO2007073486A2 (en) 2005-12-20 2007-06-28 Duke University Methods and compositions for delivering active agents with enhanced pharmacological properties
US8841255B2 (en) 2005-12-20 2014-09-23 Duke University Therapeutic agents comprising fusions of vasoactive intestinal peptide and elastic peptides
WO2008058288A2 (en) * 2006-11-09 2008-05-15 Proprius Pharmaceuticals, Inc. Sustained release methotrexate formulations and methods of use thereof
EP2111414B1 (en) 2007-02-15 2014-07-02 Indiana University Research and Technology Corporation Glucagon/glp-1 receptor co-agonists
DE602008005596D1 (en) 2007-06-21 2011-04-28 Univ Muenchen Tech BIOLOGICALLY ACTIVE PROTEINS WITH INCREASED IN VIVO AND / OR IN VITRO STABILITY
PA8830501A1 (en) 2008-06-17 2010-07-27 Univ Indiana Res & Tech Corp GLUCAGON / GLP-1 RECEIVER CO-AGONISTS
EP2307038A4 (en) 2008-06-27 2013-03-27 Univ Duke Therapeutic agents comprising elastin-like peptides
US8548622B2 (en) 2009-07-22 2013-10-01 Under Armour, Inc. Uniform compression garment and method of manufacturing garment
ES2870914T3 (en) * 2009-08-14 2021-10-28 Phasebio Pharmaceuticals Inc Modified vasoactive intestinal peptides
US20120213781A1 (en) * 2011-02-11 2012-08-23 Zyngenia, Inc. Monovalent and Multivalent Multispecific Complexes and Uses Thereof
WO2013003449A2 (en) 2011-06-27 2013-01-03 Phasebio Pharmaceuticals, Inc. Methods of treatment with glp-1 receptor agonists
AU2012346058A1 (en) 2011-11-28 2014-06-12 Phasebio Pharmaceuticals, Inc. Therapeutic agents comprising insulin amino acid sequences
US20150290328A1 (en) 2012-11-20 2015-10-15 Phasebio Pharmaceuticals, Inc. Formulations of active agents for sustained release

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080312157A1 (en) * 2005-02-11 2008-12-18 Amylin Pharmaceuticals, Inc. Gip analog and hybrid polypeptides with selectable properties

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9919032B2 (en) 2012-11-20 2018-03-20 Phasebio Pharmaceuticals, Inc. Method for administering a sustained release formulation
WO2017192449A1 (en) * 2016-05-06 2017-11-09 Phasebio Pharmaceuticals, Inc. Elp fusion proteins for controlled and sustained release
KR20190005171A (en) * 2016-05-06 2019-01-15 파세비오 파마수티컬스 인코포레이티드 ELP fusion protein for controlled sustained release
CN109310641A (en) * 2016-05-06 2019-02-05 费斯生物制药公司 For controlled and sustained release ELP fusion protein
EP4043010A1 (en) * 2016-05-06 2022-08-17 Phasebio Pharmaceuticals, Inc. Elp fusion proteins for controlled and sustained release
KR102449167B1 (en) * 2016-05-06 2022-09-28 파세비오 파마수티컬스 인코포레이티드 ELP fusion protein for controlled sustained release
KR20220136469A (en) * 2016-05-06 2022-10-07 파세비오 파마수티컬스 인코포레이티드 Elp fusion proteins for controlled and sustained release
AU2017261216B2 (en) * 2016-05-06 2023-08-03 Immunoforge Co., Ltd. ELP fusion proteins for controlled and sustained release
KR102646323B1 (en) * 2016-05-06 2024-03-13 이뮤노포지 주식회사 Elp fusion proteins for controlled and sustained release

Also Published As

Publication number Publication date
WO2014081849A1 (en) 2014-05-30
US9919032B2 (en) 2018-03-20
US20170136092A1 (en) 2017-05-18

Similar Documents

Publication Publication Date Title
US9919032B2 (en) Method for administering a sustained release formulation
CA2846209C (en) Systemic sustained release formulation comprising elastin­like peptide repeating sequence and protein active agent
EP1909824B1 (en) Pharmaceutical formulations comprising incretin peptide and aprotic polar solvent
EP3452021B1 (en) Elp fusion proteins for controlled and sustained release
KR102161177B1 (en) A liquid formulation of long acting Insulinotropic peptide conjugate
US20230414773A1 (en) Elp fusion proteins for controlled and sustained release
US20200360267A1 (en) THERAPEUTIC METHODS FOR THE TREATMENT OF DIABETES AND RELATED CONDITIONS FOR PATIENTS WITH HIGH BASELINE HbA1c
CA2856967A1 (en) Therapeutic agents comprising insulin amino acid sequences
EP2945643A1 (en) Therapeutic agents, compositions, and methods for glycemic control

Legal Events

Date Code Title Description
AS Assignment

Owner name: PHASEBIO PHARMACEUTICALS, INC., PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ARNOLD, SUSAN;PRIOR, CHRISTOPHER;GEORGOPOULOS, LYNNE;SIGNING DATES FROM 20150513 TO 20150519;REEL/FRAME:035676/0889

AS Assignment

Owner name: PHASEBIO PHARMACEUTICALS, INC., PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ARNOLD, SUSAN;PRIOR, CHRISTOPHER;GEORGOPOULOS, LYNNE;SIGNING DATES FROM 20150513 TO 20150519;REEL/FRAME:036518/0667

STCB Information on status: application discontinuation

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