US20220389071A9 - Long-lasting glp1 analogue drug for type-2 diabetes - Google Patents

Long-lasting glp1 analogue drug for type-2 diabetes Download PDF

Info

Publication number
US20220389071A9
US20220389071A9 US17/184,248 US202117184248A US2022389071A9 US 20220389071 A9 US20220389071 A9 US 20220389071A9 US 202117184248 A US202117184248 A US 202117184248A US 2022389071 A9 US2022389071 A9 US 2022389071A9
Authority
US
United States
Prior art keywords
glp1
fusion protein
glucagon
glp
disease
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.)
Pending
Application number
US17/184,248
Other versions
US20220267402A1 (en
Inventor
Yang Wei
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.)
Serpentide Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US17/184,248 priority Critical patent/US20220389071A9/en
Priority to EP21873103.2A priority patent/EP4216987A1/en
Priority to PCT/US2021/022296 priority patent/WO2022066212A1/en
Priority to CN202180001518.5A priority patent/CN114980916A/en
Publication of US20220267402A1 publication Critical patent/US20220267402A1/en
Publication of US20220389071A9 publication Critical patent/US20220389071A9/en
Assigned to SERPENTIDE INC. reassignment SERPENTIDE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WEI, YANG
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/723G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH receptor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2869Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against hormone receptors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/35Fusion polypeptide containing a fusion for enhanced stability/folding during expression, e.g. fusions with chaperones or thioredoxin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site

Definitions

  • Obesity has become a major health issue in the United States. There's more than 40% Americans are considered to be overweight or obese by modern standards due to poor diet, lack of exercise or other unhealthy behavior. Obesity is an important underlying risk factor for developing other diseases such as heart disease, stroke, and diabetes. Typically, a modest decrease in excess body weight decreases the risk of developing certain obesity-associated diseases such as heart disease and diabetes.
  • Diabetes mellitus is a disorder in which the body does not produce enough or respond normally to insulin, causing blood sugar level to be abnormally high. Long term levels of high blood sugar often result in undesirable long-term health consequences including heart disease, stroke, poor overall circulation, and in severe cases, lower limb amputation. Treatment of diabetes usually involves controlling and/or reducing blood sugar levels through a combination of regular exercise and dietary control along with certain medications such as insulin and/or metformin.
  • GLP-1 glucagon-like peptide-1 receptor agonists that target the incretin pathway.
  • GLP1 glucagon-like peptide-1
  • GLP1 exerts its main effect by stimulating glucose-dependent insulin release from the pancreatic islets. It has also been shown to slow gastric emptying, inhibit inappropriate post-meal glucagon release and reduce food intake.
  • GLP1 is quickly inactivated and/or degraded by the enzyme dipeptidyl peptidase 4 (DPP4) leading very short half-life of about 1.5 minutes. This is undesirable as it severely limits its effectiveness in regulating blood sugar levels.
  • DPP4 dipeptidyl peptidase 4
  • Longer-acting derivatives of GLP1 as well as GLP1 receptor agonists including fusion proteins comprising GLP1 provide an improved incretin effect and therefore, have been studied for diabetes control.
  • GLP1 analogues, fusion proteins and GLP1 receptor agonists are well known in the art, with exemplary examples disclosed, for example, in U.S. Pat. No.
  • GLP1 peptide variants and GLP1 receptor agonists that are resistant to degradation by DPP4 and other factors have also been explored.
  • Previous work has shown that various amino acid substitutions at position 8 of GLP1 (7-37) make such peptides more resistant to DPP4, thus conferring a longer half-life.
  • this approach still does come close to substantially fully protecting the GLP1 from DPP4 cleavage, and the middle part of the GLP1 molecule remains subject to other protease cleavage.
  • GLP1 variants and GLP1 receptor agonists are disclosed in US 2019/0091296 A1 to Wei et al. which is hereby incorporated by reference in its entirety. Although those variants provide the protection for the DPP4 cleavage, the middle part of GLP1 is still subject to be degraded by other proteases in the blood.
  • GLP1 peptide variants and GLP1 receptor agonists that are further resistant to DPP4 and other protease degradation having improved pharmacokinetic properties and having increased potency and sustained invivo activity in glycemic control.
  • GLP1 peptide variants and GLP1 receptor agonists that are further resistant to degradation having improved pharmacokinetic properties and having increased potency and sustained in vivo activity in glycemic control.
  • GLP1 peptide variants with a protection sequence comprising the Extracellular domain (ECD) of the human GLP1 receptor to protect it from protease cleavage.
  • ECD Extracellular domain
  • GLP1 peptide variants with a protection sequences that comprising an anti GLP1 antibody, preferably the Fab portions of the antibody, an nanobody or BiTE that can bind to GLP1 peptide variants and protect it from protease cleavage.
  • the glucagon-like peptide-1 receptor is a receptor protein typically found on beta cells of the pancreas and on neurons of the brain. It is involved in the control of blood sugar level by enhancing insulin secretion. In humans it is synthesized by the gene GLP1R, which is present on chromosome 6. It is a member of the glucagon receptor family of G protein-coupled receptors.
  • GLP1R includes two domains, one extracellular (ECD) that binds the C-terminal helix of GLP-1, and one transmembrane (TMD) domain that binds the N-terminal region of GLP-1.
  • ECD extracellular
  • TMD transmembrane
  • the TMD domain there is a fulcrum of polar residues that regulates the biased signaling of the receptor while the transmembrane helical boundaries and extracellular surface are a trigger for biased agonism.
  • a fusion protein wherein a receptor's extracellular domain (ECD) of the peptide, or a protection antibody (e.g., an antibody's Fab region, a nanobody or bispecific T-cell engager “BiTE” antibody in certain preferred embodiments) which binds to peptides, may be fused to the N-terminus of the peptide through a linker that contains a protease cleavage site (i.e., Factor Xa).
  • the fused receptor ECD or protection antibody may bind to the peptide and protect it from DPP4 and other protease degradation, thus conferring a longer half-life.
  • the fusion protein is, and remains inactive for the period before Factor Xa digestion removes the receptor ECD or protection antibody.
  • an hGLP1R ECD structure consistent with the present invention was linked to the N-terminus of a GLP1 analogue (e.g., Eli Lily's TrulicityTM drug although others may be used if desired) through a 3xG4S linker followed by a protease cleavage site, for example Factor Xa.
  • a GLP1 analogue e.g., Eli Lily's TrulicityTM drug although others may be used if desired
  • a 3xG4S linker followed by a protease cleavage site, for example Factor Xa.
  • the GLP1R ECD binds to GLP1, which confers two primary benefits: 1) substantially reduces or eliminates the DPP4 cleavage, and 2) protects the middle section of the GLP1 molecule from degradation due to other proteases in the blood.
  • the rate of release of the GLP-1 analogue by Factor Xa can be controlled by modifying the Factor Xa digestion sequence, a more constant GLP-1 analogue blood level may be achieved and thereby reduce undesirable side effects associated with varying blood sugar levels.
  • a point mutation e.g., R108G
  • deletion position R108 to E116
  • the fusion proteins of the present invention were tested for their ability to stimulate cAMP production in HEK cell line that stably expresses the human GLP1 receptor illustrating a significant improvement over previously known molecular structures and methods.
  • aspects of the invention are directed toward method of lowering blood sugar level by administering a pharmaceutical composition including one or more of the fusion proteins described herein in a therapeutically effective amount.
  • Such a method may be used to treat and/or prevent metabolic disorders including diabetes mellitus, obesity, insulin resistance, hypertension, dyslipidemia, Type 2 diabetes, Type 1 diabetes, prediabetes, cardiovascular disease, atherosclerosis, congestive heart failure, coronary heart disease, arteriosclerosis, peripheral artery disease, stroke, respiratory dysfunction, renal disease, fatty liver disease, non-alcoholic steatohepatitis (NASH), and metabolic syndrome.
  • metabolic disorders including diabetes mellitus, obesity, insulin resistance, hypertension, dyslipidemia, Type 2 diabetes, Type 1 diabetes, prediabetes, cardiovascular disease, atherosclerosis, congestive heart failure, coronary heart disease, arteriosclerosis, peripheral artery disease, stroke, respiratory dysfunction, renal disease, fatty liver disease, non-alcoholic steatohepatitis (NASH), and metabolic syndrome.
  • metabolic disorders including diabetes mellitus, obesity, insulin resistance, hypertension, dyslipidemia, Type 2 diabetes, Type 1 diabetes, prediabetes, cardiovascular disease, atherosclerosis, congestive heart failure
  • the methods described above may also include administering the pharmaceutical composition in combination with a second therapeutic agent or therapy which may include insulin or insulin analogue, metformin, a thiazolidinedione, a sulfonylurea, a biguanide, chlorpropamide, a glinide, an alpha glucosidase inhibitor, nateglinide, a DPP4 inhibitor, pramlintide, sitagliptin, bromocriptine, a SGLT2 inhibitor, canagliflozin, an antihypertensive drug, a statin, aspirin, dietary modification, exercise, and a dietary supplement.
  • a second therapeutic agent or therapy which may include insulin or insulin analogue, metformin, a thiazolidinedione, a sulfonylurea, a biguanide, chlorpropamide, a glinide, an alpha glucosidase inhibitor, nateglinide, a
  • FIG. 1 is a general pictorial representation of the present invention showing the general construction of embodiments of the fusion protein of the present invention.
  • FIG. 2 is a general pictorial representation of the present invention showing Type 1 molecule as GLP1 analogue is released from the protection sequence in accordance with an aspect of the present invention.
  • FIG. 3 is a general pictorial representation of the present invention showing Type 2 molecule as GLP1 analogue is released from the protection sequence in accordance with an aspect of the present invention.
  • FIG. 4 are graphs showing EC50 values of the GLP analog control molecules and molecules representing various embodiments of the of the present invention.
  • FIG. 5 is bar chart showing of the cAMP Assay showing activity of embodiments of the present after the GLP1 analogue is released during digestion.
  • FIG. 6 is graph showing the pharmacokinetic profiles of two molecules constructed in accordance with embodiments of the present invention over 7 days.
  • FIG. 7 is graph showing glucose levels of test subject in response to a molecule of one embodiment of the present invention on day 2.
  • FIG. 8 is graph showing glucose AUC of test subject in response to a molecule of one embodiment of the present invention on day 2 compared to a control.
  • FIG. 9 is graph showing glucose levels of test subject in response to a molecule of one embodiment of the present invention on day 6.
  • FIG. 10 is graph showing glucose AUC of test subject in response to a molecule of one embodiment of the present invention on day 6 compared to a control.
  • FIG. 11 a is graph illustrating IPGTT test results of diabetic test subjects in response to two molecular variants of molecules of the present invention on day 3.
  • FIG. 11 b is graph illustrating IPGTT test results of diabetic test subjects in response to two molecular variants of molecules of the present invention on day 6.
  • FIG. 11 c is graph illustrating IPGTT test results of diabetic test subjects in response to two molecular variants of molecules of the present invention on day 14.
  • TrulicityTM are fusion proteins that include two substantially identical, disulfide-linked chains, each containing an N-terminal GLP-1 analog sequence covalently linked to the Fc portion of a modified human immunoglobulin G4 (IgG4) heavy chain by a small peptide linker and is commonly produced using a mammalian cell (e.g., hamster ovary) culture.
  • IgG4 immunoglobulin G4
  • the GLP-1 analog portion of such molecules is homologous to native human GLP-1 (7-37). Structural modifications are introduced in the GLP-1 part of the molecule responsible for interaction with DPP-4. Additional modifications were made in an area with a potential T-cell epitope and in the areas of the IgG4 Fc part of the molecule responsible for binding the high-affinity Fc receptors and half-antibody formation (generally referred to herein a the “Fc part”).
  • This configuration activates the GLP-1 receptor, a membrane-bound cell-surface receptor coupled to adenylyl cyclase in pancreatic beta cells and increases intracellular cyclic AMP (cAMP) in beta cells leading to glucose-dependent insulin release with an extended half-life.
  • cAMP cyclic AMP
  • FIG. 1 A general pictorial representation 100 illustrating aspects of the fusion protein concept in accordance with the present invention is shown in FIG. 1 .
  • molecule 102 is the base GLP-1 agonist molecule which may be fused to a protection sequence 104 (the ECD of GLP1R or an antibody's Fab region, a nanobody or bispecific T-cell engager “BiTE” antibody in certain preferred embodiments) through an amino acid linker region 110 , which together may collectively form a protected GLP-1 molecule 106 .
  • a protection sequence 104 the ECD of GLP1R or an antibody's Fab region, a nanobody or bispecific T-cell engager “BiTE” antibody in certain preferred embodiments
  • protected molecule 106 may then be fused to the IgG4 Fc part 108 through an amino acid linker region (e.g., 3xG4S) to create fusion protein 114 (Type 1), or to a suitable anti GLP1R antibody 112 to produce fusion protein 116 (Type 2).
  • an amino acid linker region e.g., 3xG4S
  • fusion protein 114 Type 1
  • suitable anti GLP1R antibody 112 to produce fusion protein 116 (Type 2).
  • any suitable GLP-1 agonist may be used for molecule 102 if desired, such as any Glucagon/secretin superfamily molecules which may include, but is not limited to, Glucagon, GLP-1, GLP-2, GIP, VIP, PACAP, Exendin-4, GLP1/GIP dual agonist and GLP1/GIP/Glucagon triple agonist, or other suitable peptide drugs, etc.
  • the present invention may include a GLP1 variant comprising a GLP1R ECD fused to the N-terminal of Trulicity though a 3xG4S linker plus a Factor Xa protease digestion site.
  • the present invention may include the fusion protein of Embodiment 1, wherein the GLP1 ECD contains a mutation at the position of 108 , from Arg to Gly, or to other amino acids to eliminate Factor Xa cleavage site.
  • the present invention may include the fusion protein of Embodiment 1, wherein the GLP1 ECD contains a 9 amino acids deletion at the C-terminus from position 108 to 116 (RGERSSPEE), in order to remove Factor Xa cleavage site.
  • GLP1 ECD contains a 9 amino acids deletion at the C-terminus from position 108 to 116 (RGERSSPEE), in order to remove Factor Xa cleavage site.
  • the present invention may include a GLP1 variant comprising a Fab part of an anti GLP1R antibody, which is fused to the N-terminal of Trulicity through a 3xG4S linker plus a Factor Xa protease digestion site.
  • the present invention may include a GLP1 variant comprising a GLP1 protection nanobody fused to the N-terminal of Trulicity through a 3xG4S linker plus a Factor Xa protease digestion site.
  • the present invention may include a GLP1 variant comprising an anti GLP1 antibody in BiTE format fused to the N-terminal of Trulicity through a 3xG4S linker plus a Factor Xa protease digestion site.
  • the present invention may include the fusion proteins described in any of Embodiment 1 to 6, wherein the linker is 1xG4S, 2xG4S or 3xG4S plus the Factor Xa digestion site or its variant (i.e., RKRR, RGER, RKR, RR etc.), or other suitable protease digestion sites.
  • the linker is 1xG4S, 2xG4S or 3xG4S plus the Factor Xa digestion site or its variant (i.e., RKRR, RGER, RKR, RR etc.), or other suitable protease digestion sites.
  • the present invention may include the fusion proteins of any Embodiment 1 to 7, wherein the GLP1 part of Trulicity is select from one of the Glucagon/secretin superfamily includes, but not limited to: Glucagon, GLP-1, GLP-2, GIP, VIP, PACAP, Exendin-4, GLP1/GIP dual agonist and GLP1/GIP/Glucagon triple agonist.
  • the present invention may include the fusion proteins described in any of Embodiment 1 to 8, wherein the Fc part of Trulicity is an anti GLP1R antibody, or an antibody against the receptor corresponding to the agonist has been used, or may be a bispecific antibody if a dual GLP1/GIP agonist is used.
  • a protected fusion protein 106 wherein a receptor's extracellular domain (ECD) of the peptide or a protection antibody 104 which binds to peptides 102 through linker 110 that contains a protease cleavage site (e.g., Factor Xa), may be fused to the N-terminus of the GLP1 variant fused to human Fc (e.g., Trulicity) to form Type 1 fusion protein 114 , or a GLP1 variant fused to an antibody 112 to form Type 2 fusion protin 116 .
  • ECD extracellular domain
  • a protection antibody 104 which binds to peptides 102 through linker 110 that contains a protease cleavage site (e.g., Factor Xa)
  • Fc e.g., Trulicity
  • the receptor ECD or protection antibody 104 may bind to the peptide 102 and protect it from DPP4 and other protease degradation, thus conferring a longer half-life.
  • the fusion protein is, and remains inactive for the period before digestion removes receptor ECD or protection antibody 104 .
  • Type 1 molecule 214 constructed in accordance with the present invention, wherein the GLP1R ECD 204 was linked to the N-terminus of a GLP-1 analogue 208 (e.g., Eli Lily's TrulicityTM drug although others such as OzempicTM may be used if desired) through a linker 210 (3xG4S followed by a protease cleavage site (e.g., Factor Xa)).
  • GLP-1 analogue 208 e.g., Eli Lily's TrulicityTM drug although others such as OzempicTM may be used if desired
  • linker 210 3xG4S followed by a protease cleavage site (e.g., Factor Xa)).
  • protease cleavage site e.g., Factor Xa
  • the GLP1R ECD binds to GLP1, which confers two primary benefits: 1) substantially reduces or eliminates the DPP4 cleavage, and 2) protects the middle section of the GLP1 molecule from degradation due to other proteases in the blood.
  • FIG. 2 shows from left to right hGLP1R ECD protected Type 1 molecule 214 shedding protection sequence 204 in a controlled in predetermined manner as digestion progresses producing a released GLP1-hFc(Trulicity) molecule that provides users with substantially constant drug concentration as shown in FIGS. 4 - 11 discussed below.
  • FIG. 3 a more particularized version of the Type 2 molecule of FIG. 1 is depicted.
  • an hGLP1 antibody protected Type 2 molecule 314 constructed in accordance with the present invention.
  • a protection sequence 304 e.g., hGLP1R ECD or an anti GLP1 antibody
  • 3xG4S linker 310 contains a protease cleavage site (e.g., Factor Xa) to form molecule 306
  • molecule 306 was linked to the N-terminus of the light chain of an anti-GLP1R targeting antibody to form fusion protein 314 .
  • the GLP1R ECD binds to GLP1, which, as above in FIG. 3 confers two primary benefits: 1) substantially reduces or eliminates the DPP4 cleavage, and 2) protects the middle section of the GLP1 molecule from degradation due to other proteases in the blood.
  • FIG. 3 shows from left to right the Type 2 molecule 314 shedding protection sequence 304 in a controlled in predetermined manner as digestion progresses producing a released GLP1-antibody molecule 318 that provides users with substantially constant blood drug concentration as shown in the charts below.
  • a hGLP1R ECD molecule (SEQ ID NO 3) was linked to the N-terminus of Trulicity through a linker contains a 3xG4S and a protease cleavage site (Factor Xa, NEB lot#P8010S).
  • the plate was coated with Mouse anti-human IgG (Fc) (CELLWAYLAB lot#C010202) at 4 ug/ml, 4° C. overnight, block with 0.2% BSA/0.1%PC/0.1% at room temperate for one hour, then samples prepared from mouse serum were added and incubated at room temperate for an hour.
  • the Trulicity is used as the standard curve.
  • Anti GLP-1 antibody (ThermoFisher, Catalog#: ABS03310B005) is used for detecting active Trulicity.
  • the reaction was stopped by 2M H2SO4, and read at 450nm with plate reader (CMax Plus, Molecular Devices).
  • mice received a single subcutaneous injection of hFc control pTG1, p9, p11, and Trulicity at 200 nmol/kg.
  • Intraperitoneal glucose tolerance tests IPGTT were performed on Day 3, Day 6 and 14 after overnight fasting with blood glucose measurements at 0, 15, 30, 60, and 120 minutes.
  • Mean ⁇ SEM of blood glucose levels at each time point and glucose area under curve (AUC) were calculated for each group and shown in FIGS. 7 - 11 .
  • Excel t-test was used to assess the significance to the control group, *: p ⁇ 0.05, **: p ⁇ 0.01, ***: p ⁇ 0.001, ****: p ⁇ 0.0001
  • TrulicityTM, and the released p7, p9 and p11 show EC50 values of 22 nM, 25 nM, 17.6 nM and 26.9 nM, respectively, for GLP1R activation.
  • the EC50 for released pTG3 and p10 is only 617 nM and 120 nM, indicating the Factor Xa digestion were not at predicted sites. This was confirmed by N-terminal amino acids sequence analysis.
  • the EC50 for p8 is undetectable because there is no Factor Xa digestion site, therefore the active Trulicity cannot be released.
  • the p7, p9, p10 and p11 only show activities after Factor Xa digestion (released).
  • the p8 (SEQ ID8) negative control shows no cAMP activity because it contains no Factor Xa digestion site and the active Trulicity could't be released.
  • GLP1 fusion proteins EC50 hGLPIR ECD-3xG4S-RKRR-Trulicity(pTG3, 617 nM lot#U5585DH140S05/P90011809) hGLPIR ECD(R108G)-3xG4S-RGER-Trulicity(p7, 25 nM lot#U2243DL140-4/P9EA001) hGLPIR ECD(R108G)-3xG4S Trulicity(p8, lot# undetectable U2243DL140-9/P9E A001) hGLPIR ECD(1-116)-3xG4S-RGER-Trulicity(p9, lot 17.6 nM U2243DL140-14/P9E A001) hGLPIR ECD(R108G)-3xG4S-RR-Trulicity(p10, lot# 120 nM U2243DL140-19/P9E A001) hGLPIR ECD(RCD(R108G)-3xG4S-RR
  • FIG. 6 shows Pharmacokinetic profiles of released p9 and pll in C57/BL Mice after subcutaneous (SC) administration of 200 nmol/kg.
  • the half-life for released p11 is 6 days, the half-life for p9 is longer than 7 days because 75% of released p9 is still detected at day 7.
  • Both proteins show relative constant blood drug concentration from day 1 to day 7.
  • FIGS. 7 - 10 show favorable IPGTT results in C57/BL mice, that is, a single administration of p11 variant of the present invention provided significant glucose reductions at day 2 and 6.
  • FIGS. 11 a , 11 b , and 11 c show the IPGTT results in diabetic BKS/DB mice, that is, a single administration of p9 and pll variants of the invention provided significant glucose reductions at day 3, day 6 and day 14.
  • the GLP1 receptor agonists of the present invention may include proteins having amino acid sequences that may vary from those of the described GLP1 receptor agonists, but that retain the ability to bind GLP1 receptor. Such variant GLP1 receptor agonists may comprise one or more additions, deletions, or substitutions of amino acids when compared to parent sequence but exhibit biological activity that is essentially equivalent to that of the described GLP1 receptor agonists.
  • the GLP1 receptor agonist-encoding DNA sequences of the present invention may encompass sequences that comprise one or more additions, deletions, or substitutions of nucleotides when compared to the disclosed sequence, but that encode a GLP1 receptor agonist that is essentially bioequivalent to a GLP1 receptor agonist of the invention.
  • Two proteins may be considered bioequivalent if, for example, they are pharmaceutical equivalents or pharmaceutical alternatives whose rate and extent of absorption are substantially the same do not exhibit a significant difference when administered at the same molar dose under similar experimental conditions, which may include either single dose or multiple doses. Some proteins will be considered equivalents or pharmaceutical alternatives if they are equivalent in the extent of their absorption but not in their rate of absorption and yet may be considered bioequivalent because such differences in the rate of absorption are intentional and are reflected in the labeling, are not essential to the attainment of effective body drug concentrations on, e.g., chronic use, and are considered medically insignificant for the particular drug product studied.
  • two GLP1 receptor agonist proteins are bioequivalent if there are no clinically meaningful differences in their safety, purity, potency or efficacy.
  • two GLP1 receptor agonist proteins are bioequivalent if a patient can be switched one or more times between the reference product and the biological product without an expected increase in the risk of adverse effects, including a clinically significant change in immunogenicity, or diminished effectiveness, as compared to continued therapy without such switching.
  • two GLP1 receptor agonist proteins are bioequivalent if they both act by a common mechanism or mechanisms of action for the condition or conditions of use, to the extent that such mechanisms are known.
  • Bioequivalence may be demonstrated by in vivo and/or in vitro methods.
  • Bioequivalence measures include, e.g., (a) an in vivo test in humans or other mammals, in which the concentration of the protein or its metabolites is measured in blood, plasma, serum, or other biological fluid as a function of time; (b) an in vitro test that has been correlated with and is reasonably predictive of human in vivo bioavailability data; (c) an in vivo test in humans or other mammals in which the appropriate acute pharmacological effect of the protein (or its target) is measured as a function of time; and (d) in a well-controlled clinical trial that establishes safety, efficacy, or bioavailability or bioequivalence of an antigen-binding protein.
  • the invention provides therapeutic compositions comprising the GLP1 receptor agonists.
  • Therapeutic compositions in accordance with the invention will be administered with suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • a multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. “Compendium of excipients for parenteral formulations” PDA (1998) J Pharm Sci Technol 52:238-311.
  • the dose of GLP1 receptor agonist may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like.
  • an antigen-binding protein of the present invention is used for treating a disease or disorder in an adult patient, or for preventing such a disease, it is advantageous to administer the antigen-binding protein of the present invention normally at a single dose of about 0.001 to about 100 mg/kg body weight, more preferably about 0.001 to about 60, about 0.01 to about 10, or about 0.01 to about 1 mg/kg body weight.
  • the frequency and the duration of the treatment can be adjusted.
  • the antigen-binding protein or antigen-binding fragment thereof of the invention can be administered as an initial dose of at least about 0.001 mg to about 100 mg, about 0.001 to about 50 mg, about 0.005 to about 50 mg, about 0.01 to about 40 mg, to about 30 mg, or to about 10 mg.
  • the initial dose may be followed by administration of a second or a plurality of subsequent doses of the GLP1 receptor agonist in an amount that can be approximately the same or less than that of the initial dose, wherein the subsequent doses are separated by at least 1 day to 3 days; at least one week, at least 2 weeks; at least 3 weeks; at least 4 weeks; at least 5 weeks; at least 6 weeks; at least 7 weeks; at least 8 weeks; at least 9 weeks; at least 10 weeks; at least 12 weeks; or at least 14 weeks.
  • Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al. (1987) J. Biol. Chem. 262:4429-4432).
  • Methods of introduction include, but are not limited to, intradermal, transdermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural and oral routes.
  • the composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • the pharmaceutical composition can be also delivered in a vesicle, in particular a liposome (see, for example, Langer (1990) Science 249:1527-1533).
  • Nanoparticles to deliver the GLP1 receptor agonists of the present invention is also contemplated herein.
  • Protein-conjugated nanoparticles may be used both for therapeutic and diagnostic applications.
  • Nanoparticles may be developed and conjugated to antigen-binding proteins contained in pharmaceutical compositions to target cells. Nanoparticles for drug delivery have also been described in, for example, U.S. Pat. No. 8,257,740, or U.S. Pat. No. 8,246,995, each incorporated herein in its entirety.
  • the pharmaceutical compositions or treatments of the present invention may be delivered in a controlled release system.
  • a pump may be used.
  • polymeric materials can be used.
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose.
  • the injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous, intracranial, intraperitoneal and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antigen-binding protein or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • a nonionic surfactant e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil
  • oily medium there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • a pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe.
  • a pen or wand type delivery device readily has applications in delivering a pharmaceutical composition of the present invention.
  • Such a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused.
  • a disposable pen delivery device there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention.
  • Examples include, but certainly are not limited to AUTOPENTM (Owen Mumford, Inc., Woodstock, UK), DISETRONICTM pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25TM pen, HUMALOGTM pen, HUMALIN 70/30TM pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPENTM I, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIORTM (Novo Nordisk, Copenhagen, Denmark), BDTM pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPENTM, OPTIPEN PROTM, OPTIPEN STARLETTM, and OPTICLIKTM (Sanofi-Aventis, Frankfurt, Germany), to name only a few.
  • AUTOPENTM Owen Mumford, Inc., Woodstock, UK
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • the amount of the GLP1 receptor agonist contained is generally about 0.001 to about 100 mg per dosage form in a unit dose; especially in the form of injection, it is preferred that the GLP1 receptor agonist is contained in about 0.001 to about 100 mg and in about 0.01 to about 100 mg for the other dosage forms.
  • the GLP1 receptor agonists of the invention are useful to treat subjects that are overweight, obese and/or prevent or treat one or more obesity-associated disorders such as heart disease, stroke, and diabetes.
  • the GLP1 receptor agonists of the invention are useful to treat subjects suffering from diabetes and/or prevent one or more complications of associated with diabetes such as heart disease, stroke, kidney disease, retinopathy, blindness and peripheral nerve damage.
  • one or more GLP1 receptor agonist fusion proteins of the present invention may be used as a preventive measure for patients in danger of developing diabetes (e.g., from type 2 diabetes).
  • risks include, but are not limited to, patients of advanced age, pregnant women, and/or other risk factors including family history of obesity, high blood cholesterol, smoking, excessive alcohol consumption, and/or lack of exercise.
  • the proteins of the invention may be used for the preparation of a pharmaceutical composition or medicament for treating patients suffering from a disease or disorder such as diabetes and obesity.
  • the GLP1 receptor agonists may be used as complimentary or supplemental therapy with any other suitable therapy known to those skilled in the art useful for treating or ameliorating a disease or disorder associated with hyperglycemia such as diabetes (e.g., type 2 diabetes).
  • Combination therapies contemplated by the present invention may include a GLP1 receptor agonist of the invention and any suitable additional therapeutic agent that may be advantageously combined therewith, such as GLP1 receptor agonist of the invention, or a biologically active fragment of the invention as would be appreciated by one of ordinary skill in the art. Further combination may include the GLP1 receptor agonists of the present inventio combined synergistically with one or more drugs or therapy used to treat any disease or disorder associated with hyperglycemia (e.g., diabetes). In some embodiments, the GLP1 receptor agonists of the invention may be combined with one or more other therapeutic agent(s) to reduce blood sugar levels in a subject, or to ameliorate one or more symptoms of diabetes.
  • the GLP1 receptor agonists of the present invention may be used in combination with an insulin (insulin or an insulin analog), insulin sensitizers such as biguanides (e.g., metformin), and thiazolidinediones (e.g., rosiglitazone), insulin secretagogues such as sulphonylureas (e.g., chlorpropamide), and glinides (e.g., nateglinide), alpha-glucosidase inhibitors (e.g., acarbose), dipeptidyl peptidase 4 (DPP4) inhibitors (e.g., sitagliptin), pramlinitide, bromocriptine, sodium glucose cotransporter 2 (SGLT-2) inhibitors (e.g., canagliflozin), an anti-hypertensive drug (e.g., an angiotensin-converting enzyme inhibitor, an angiotensin receptor blocker, a diuretic, a calcium
  • the GLP1 receptor agonists may be administered in combination with a one or more therapeutic agent(s) including: insulin, an insulin analog, metformin, rosiglitazone, pioglitazone, chlorpropamide, glibenclamide, glimepiride, glipizide, tolazamide, tolbutamide, nateglinide, repaglinide, acarbose, miglitol, exenatide, liraglutide, albiglutide, dulaglutide, sitagliptin, saxagliptin, linagliptin, alogliptin, pramlinitide, bromocriptine quick-release, canagliflozin, dapagliflozin, empagliflozin, diet modifications and exercise.
  • a one or more therapeutic agent(s) including: insulin, an insulin analog, metformin, rosiglitazone, pioglitazone, chlorpropamide, glibenclam
  • the term “in combination with” means that additional therapeutically active component(s) may be administered prior to, concurrent with, or after the administration of the GLP1 receptor agonist of the present invention.
  • the term “in combination with” also includes sequential or concomitant administration of a GLP1 receptor agonist and a second therapeutic agent.
  • a first component may be deemed to be administered “after” a second component if the first component is administered 1 minute after, 5 minutes after, 10 minutes after, 15 minutes after, 30 minutes after, 1 hour after, 2 hours after, 3 hours after, 4 hours after, 5 hours after, 6 hours after, 12 hours after, 24 hours after, 36 hours after, 48 hours after, 60 hours after, 72 hours after administration of the second component.
  • the additional therapeutically active component(s) may be administered to a subject concurrent with administration of a GLP1 receptor agonist of the present invention.
  • Constant administration includes, e.g., administration of a GLP1 receptor agonist and an additional therapeutically active component to a subject in a single dosage form, or in separate dosage forms administered to the subject within about 30 minutes or less of each other. If administered in separate dosage forms, each dosage form may be administered via the same route (e.g., both the GLP1 receptor agonist and the additional therapeutically active component may be administered intravenously, etc.); alternatively, each dosage form may be administered via a different route (e.g., the GLP1 receptor agonist may be administered intravenously, and the additional therapeutically active component may be administered orally).
  • administering the components in a single dosage from, in separate dosage forms by the same route, or in separate dosage forms by different routes are all considered “concurrent administration,” for purposes of the present disclosure.
  • administration of a GLP1 receptor agonist “prior to”, “concurrent with,” or “after” (as those terms are defined herein above) administration of an additional therapeutically active component is considered administration of a GLP1 receptor agonist “in combination with” an additional therapeutically active component.
  • the present invention includes pharmaceutical compositions in which a GLP1 receptor agonist of the present invention is co-formulated with one or more of the additional therapeutically active component(s) as described elsewhere herein.

Abstract

Modified glucagon-like peptide (GLP1) fusion proteins with modified GLP1 polypeptides and related methods of use are described. Aspects of the disclosure further relate to fusion proteins that are GLP1 receptor agonists with a modified GLP1 fused to a stabilizing domain such as an extra cellular domain or antibody. Fusion proteins with modified GLP1 that are useful for treating or ameliorating a symptom or indication of a blood sugar disorder such as obesity and diabetes are also provided.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application claims the benefit of U.S. Provisional Application 63/081,363 filed Sep. 22, 2020 which is hereby incorporated by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • Obesity has become a major health issue in the United States. There's more than 40% Americans are considered to be overweight or obese by modern standards due to poor diet, lack of exercise or other unhealthy behavior. Obesity is an important underlying risk factor for developing other diseases such as heart disease, stroke, and diabetes. Typically, a modest decrease in excess body weight decreases the risk of developing certain obesity-associated diseases such as heart disease and diabetes.
  • Diabetes mellitus is a disorder in which the body does not produce enough or respond normally to insulin, causing blood sugar level to be abnormally high. Long term levels of high blood sugar often result in undesirable long-term health consequences including heart disease, stroke, poor overall circulation, and in severe cases, lower limb amputation. Treatment of diabetes usually involves controlling and/or reducing blood sugar levels through a combination of regular exercise and dietary control along with certain medications such as insulin and/or metformin.
  • One of the approaches used for treating diabetes and for glycemic control involves the use of glucagon-like peptide (GLP)-1 receptor agonists that target the incretin pathway. Glucagon-like peptide (GLP)-1 is a peptide hormone secreted by intestinal enteroendocrine cells. GLP1 exerts its main effect by stimulating glucose-dependent insulin release from the pancreatic islets. It has also been shown to slow gastric emptying, inhibit inappropriate post-meal glucagon release and reduce food intake.
  • However, GLP1 is quickly inactivated and/or degraded by the enzyme dipeptidyl peptidase 4 (DPP4) leading very short half-life of about 1.5 minutes. This is undesirable as it severely limits its effectiveness in regulating blood sugar levels. Longer-acting derivatives of GLP1 as well as GLP1 receptor agonists including fusion proteins comprising GLP1, provide an improved incretin effect and therefore, have been studied for diabetes control. Such GLP1 analogues, fusion proteins and GLP1 receptor agonists are well known in the art, with exemplary examples disclosed, for example, in U.S. Pat. No. 9,409,966, and published applications including US20160194371, US20170114115, US20170112904, US20160361390, US20150259416, WO2017074715, WO2016127887, EP3034514, EP2470198, and EP2373681.
  • GLP1 peptide variants and GLP1 receptor agonists that are resistant to degradation by DPP4 and other factors have also been explored. Previous work has shown that various amino acid substitutions at position 8 of GLP1 (7-37) make such peptides more resistant to DPP4, thus conferring a longer half-life. However, this approach still does come close to substantially fully protecting the GLP1 from DPP4 cleavage, and the middle part of the GLP1 molecule remains subject to other protease cleavage.
  • Certain such GLP1 variants and GLP1 receptor agonists are disclosed in US 2019/0091296 A1 to Wei et al. which is hereby incorporated by reference in its entirety. Although those variants provide the protection for the DPP4 cleavage, the middle part of GLP1 is still subject to be degraded by other proteases in the blood.
  • Accordingly, there is a need to develop GLP1 peptide variants and GLP1 receptor agonists that are further resistant to DPP4 and other protease degradation having improved pharmacokinetic properties and having increased potency and sustained invivo activity in glycemic control.
  • It therefore would be desirable to provide GLP1 peptide variants and GLP1 receptor agonists that are further resistant to degradation having improved pharmacokinetic properties and having increased potency and sustained in vivo activity in glycemic control.
  • It therefore would be desirable to provide GLP1 peptide variants with a protection sequence comprising the Extracellular domain (ECD) of the human GLP1 receptor to protect it from protease cleavage.
  • It therefore would be desirable to provide GLP1 peptide variants with a protection sequences that comprising an anti GLP1 antibody, preferably the Fab portions of the antibody, an nanobody or BiTE that can bind to GLP1 peptide variants and protect it from protease cleavage.
  • It therefore would be desirable to provide the protected GLP1 peptide variants with an anti-GLP1R targeting antibody having increased potency and sustained invivo activity in glycemic control.
  • SUMMARY OF THE INVENTION
  • One aspect of the present invention provides molecular technology for the protection of peptide-based drugs. The glucagon-like peptide-1 receptor (GLP1R) is a receptor protein typically found on beta cells of the pancreas and on neurons of the brain. It is involved in the control of blood sugar level by enhancing insulin secretion. In humans it is synthesized by the gene GLP1R, which is present on chromosome 6. It is a member of the glucagon receptor family of G protein-coupled receptors.
  • GLP1R includes two domains, one extracellular (ECD) that binds the C-terminal helix of GLP-1, and one transmembrane (TMD) domain that binds the N-terminal region of GLP-1. In the TMD domain, there is a fulcrum of polar residues that regulates the biased signaling of the receptor while the transmembrane helical boundaries and extracellular surface are a trigger for biased agonism.
  • According to one embodiment of the present invention, a fusion protein is provided wherein a receptor's extracellular domain (ECD) of the peptide, or a protection antibody (e.g., an antibody's Fab region, a nanobody or bispecific T-cell engager “BiTE” antibody in certain preferred embodiments) which binds to peptides, may be fused to the N-terminus of the peptide through a linker that contains a protease cleavage site (i.e., Factor Xa). With this approach, the fused receptor ECD or protection antibody may bind to the peptide and protect it from DPP4 and other protease degradation, thus conferring a longer half-life. In such embodiments, the fusion protein is, and remains inactive for the period before Factor Xa digestion removes the receptor ECD or protection antibody.
  • In one embodiment, an hGLP1R ECD structure consistent with the present invention was linked to the N-terminus of a GLP1 analogue (e.g., Eli Lily's Trulicity™ drug although others may be used if desired) through a 3xG4S linker followed by a protease cleavage site, for example Factor Xa. In such a fusion protein, the GLP1R ECD binds to GLP1, which confers two primary benefits: 1) substantially reduces or eliminates the DPP4 cleavage, and 2) protects the middle section of the GLP1 molecule from degradation due to other proteases in the blood.
  • Because the rate of release of the GLP-1 analogue by Factor Xa can be controlled by modifying the Factor Xa digestion sequence, a more constant GLP-1 analogue blood level may be achieved and thereby reduce undesirable side effects associated with varying blood sugar levels.
  • One particular way this may be accomplished includes certain modifications to portions of the Factor Xa digestion sequence (e.g., RKRR, RGER, RKR, RR etc.). Further, a point mutation (e.g., R108G) or deletion (position R108 to E116) may also be made in human GLP1R ECD in if desired to remove an internal Factor Xa cleavage site.
  • Before and after Factor Xa digestion, the fusion proteins of the present invention were tested for their ability to stimulate cAMP production in HEK cell line that stably expresses the human GLP1 receptor illustrating a significant improvement over previously known molecular structures and methods.
  • Other aspects of the invention are directed toward method of lowering blood sugar level by administering a pharmaceutical composition including one or more of the fusion proteins described herein in a therapeutically effective amount.
  • Such a method may be used to treat and/or prevent metabolic disorders including diabetes mellitus, obesity, insulin resistance, hypertension, dyslipidemia, Type 2 diabetes, Type 1 diabetes, prediabetes, cardiovascular disease, atherosclerosis, congestive heart failure, coronary heart disease, arteriosclerosis, peripheral artery disease, stroke, respiratory dysfunction, renal disease, fatty liver disease, non-alcoholic steatohepatitis (NASH), and metabolic syndrome.
  • The methods described above may also include administering the pharmaceutical composition in combination with a second therapeutic agent or therapy which may include insulin or insulin analogue, metformin, a thiazolidinedione, a sulfonylurea, a biguanide, chlorpropamide, a glinide, an alpha glucosidase inhibitor, nateglinide, a DPP4 inhibitor, pramlintide, sitagliptin, bromocriptine, a SGLT2 inhibitor, canagliflozin, an antihypertensive drug, a statin, aspirin, dietary modification, exercise, and a dietary supplement.
  • DESCRIPTION OF THE DRAWINGS
  • The above and other objects and advantages of the present invention will be apparent upon consideration of the following detailed description, taken in conjunction with the accompanying drawings, in which like reference characters refer to like parts throughout, and in which:
  • FIG. 1 is a general pictorial representation of the present invention showing the general construction of embodiments of the fusion protein of the present invention.
  • FIG. 2 is a general pictorial representation of the present invention showing Type 1 molecule as GLP1 analogue is released from the protection sequence in accordance with an aspect of the present invention.
  • FIG. 3 is a general pictorial representation of the present invention showing Type 2 molecule as GLP1 analogue is released from the protection sequence in accordance with an aspect of the present invention.
  • FIG. 4 are graphs showing EC50 values of the GLP analog control molecules and molecules representing various embodiments of the of the present invention.
  • FIG. 5 is bar chart showing of the cAMP Assay showing activity of embodiments of the present after the GLP1 analogue is released during digestion.
  • FIG. 6 is graph showing the pharmacokinetic profiles of two molecules constructed in accordance with embodiments of the present invention over 7 days.
  • FIG. 7 is graph showing glucose levels of test subject in response to a molecule of one embodiment of the present invention on day 2.
  • FIG. 8 is graph showing glucose AUC of test subject in response to a molecule of one embodiment of the present invention on day 2 compared to a control.
  • FIG. 9 is graph showing glucose levels of test subject in response to a molecule of one embodiment of the present invention on day 6.
  • FIG. 10 is graph showing glucose AUC of test subject in response to a molecule of one embodiment of the present invention on day 6 compared to a control.
  • FIG. 11 a is graph illustrating IPGTT test results of diabetic test subjects in response to two molecular variants of molecules of the present invention on day 3.
  • FIG. 11 b is graph illustrating IPGTT test results of diabetic test subjects in response to two molecular variants of molecules of the present invention on day 6.
  • FIG. 11 c is graph illustrating IPGTT test results of diabetic test subjects in response to two molecular variants of molecules of the present invention on day 14.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The following contains exemplary descriptions of methods and compounds in accordance with aspects of the present inventions. it will be to be understood, however, that the inventions herein are not limited to particular methods, and experimental conditions described, as such methods and conditions may vary. It is also will be understood that the terminology used herein is for the purpose of describing certain embodiments only, and is not intended to be limiting, but merely as a means for the expression of novel concepts of the invention and that the scope of the present invention will be limited only by the appended claims.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood at the time of filing by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference in their entirety including, but not limited to, U.S. Patent Publication 2019/0091296 A1 to Wei et al. which sets forth, among other things, commonly used definitions and sets forth commonly understood state of the art terminology and understanding which are used consistently herein including concepts of what is accepted understanding of bioequivalence.
  • It is an object of the invention to improve the ability of a GLP-1 analogue type drugs to resist digestion-based degradation and thereby provide longer lasting effective glucose management in users. Certain known GLP-1 receptor agonists such as Trulicity™ are fusion proteins that include two substantially identical, disulfide-linked chains, each containing an N-terminal GLP-1 analog sequence covalently linked to the Fc portion of a modified human immunoglobulin G4 (IgG4) heavy chain by a small peptide linker and is commonly produced using a mammalian cell (e.g., hamster ovary) culture.
  • The GLP-1 analog portion of such molecules is homologous to native human GLP-1 (7-37). Structural modifications are introduced in the GLP-1 part of the molecule responsible for interaction with DPP-4. Additional modifications were made in an area with a potential T-cell epitope and in the areas of the IgG4 Fc part of the molecule responsible for binding the high-affinity Fc receptors and half-antibody formation (generally referred to herein a the “Fc part”). This configuration activates the GLP-1 receptor, a membrane-bound cell-surface receptor coupled to adenylyl cyclase in pancreatic beta cells and increases intracellular cyclic AMP (cAMP) in beta cells leading to glucose-dependent insulin release with an extended half-life.
  • A general pictorial representation 100 illustrating aspects of the fusion protein concept in accordance with the present invention is shown in FIG. 1 . As shown, molecule 102 is the base GLP-1 agonist molecule which may be fused to a protection sequence 104(the ECD of GLP1R or an antibody's Fab region, a nanobody or bispecific T-cell engager “BiTE” antibody in certain preferred embodiments) through an amino acid linker region 110, which together may collectively form a protected GLP-1 molecule 106. In operation, protected molecule 106 may then be fused to the IgG4 Fc part 108 through an amino acid linker region (e.g., 3xG4S) to create fusion protein 114 (Type 1), or to a suitable anti GLP1R antibody 112 to produce fusion protein 116 (Type 2).
  • It will be understood that any suitable GLP-1 agonist may be used for molecule 102 if desired, such as any Glucagon/secretin superfamily molecules which may include, but is not limited to, Glucagon, GLP-1, GLP-2, GIP, VIP, PACAP, Exendin-4, GLP1/GIP dual agonist and GLP1/GIP/Glucagon triple agonist, or other suitable peptide drugs, etc.
  • One of ordinary skill will readily understand and recognize that using the base construct and elements described above and illustrated in FIG. 1 , at least dozens of variants of the fusion protein of the present invention may be produced if desired.
  • For example, in an Embodiment 1, the present invention may include a GLP1 variant comprising a GLP1R ECD fused to the N-terminal of Trulicity though a 3xG4S linker plus a Factor Xa protease digestion site.
  • In an Embodiment 2, the present invention may include the fusion protein of Embodiment 1, wherein the GLP1 ECD contains a mutation at the position of 108, from Arg to Gly, or to other amino acids to eliminate Factor Xa cleavage site.
  • In an Embodiment 3, the present invention may include the fusion protein of Embodiment 1, wherein the GLP1 ECD contains a 9 amino acids deletion at the C-terminus from position 108 to 116 (RGERSSPEE), in order to remove Factor Xa cleavage site.
  • In an Embodiment 4, the present invention may include a GLP1 variant comprising a Fab part of an anti GLP1R antibody, which is fused to the N-terminal of Trulicity through a 3xG4S linker plus a Factor Xa protease digestion site.
  • In an Embodiment 5, the present invention may include a GLP1 variant comprising a GLP1 protection nanobody fused to the N-terminal of Trulicity through a 3xG4S linker plus a Factor Xa protease digestion site.
  • In an Embodiment 6, the present invention may include a GLP1 variant comprising an anti GLP1 antibody in BiTE format fused to the N-terminal of Trulicity through a 3xG4S linker plus a Factor Xa protease digestion site.
  • In an Embodiment 7, the present invention may include the fusion proteins described in any of Embodiment 1 to 6, wherein the linker is 1xG4S, 2xG4S or 3xG4S plus the Factor Xa digestion site or its variant (i.e., RKRR, RGER, RKR, RR etc.), or other suitable protease digestion sites.
  • In an Embodiment 8, the present invention may include the fusion proteins of any Embodiment 1 to 7, wherein the GLP1 part of Trulicity is select from one of the Glucagon/secretin superfamily includes, but not limited to: Glucagon, GLP-1, GLP-2, GIP, VIP, PACAP, Exendin-4, GLP1/GIP dual agonist and GLP1/GIP/Glucagon triple agonist.
  • In an Embodiment 9, the present invention may include the fusion proteins described in any of Embodiment 1 to 8, wherein the Fc part of Trulicity is an anti GLP1R antibody, or an antibody against the receptor corresponding to the agonist has been used, or may be a bispecific antibody if a dual GLP1/GIP agonist is used.
  • As shown in FIG. 1 , a protected fusion protein 106 is provided wherein a receptor's extracellular domain (ECD) of the peptide or a protection antibody 104 which binds to peptides 102 through linker 110 that contains a protease cleavage site (e.g., Factor Xa), may be fused to the N-terminus of the GLP1 variant fused to human Fc (e.g., Trulicity) to form Type 1 fusion protein 114, or a GLP1 variant fused to an antibody 112 to form Type 2 fusion protin 116. Accordingly, the receptor ECD or protection antibody 104 may bind to the peptide 102 and protect it from DPP4 and other protease degradation, thus conferring a longer half-life. In such embodiments, the fusion protein is, and remains inactive for the period before digestion removes receptor ECD or protection antibody 104.
  • As shown in FIG. 2 , a more particularized version of the Type 1 molecule of FIG. 1 is depicted. As shown, a Type 1 molecule 214 constructed in accordance with the present invention, wherein the GLP1R ECD 204 was linked to the N-terminus of a GLP-1 analogue 208 (e.g., Eli Lily's Trulicity™ drug although others such as Ozempic™ may be used if desired) through a linker 210 (3xG4S followed by a protease cleavage site (e.g., Factor Xa)). These include SEQ IDs 6, 7, 8, 9, 10 and 11. In such a fusion protein, the GLP1R ECD binds to GLP1, which confers two primary benefits: 1) substantially reduces or eliminates the DPP4 cleavage, and 2) protects the middle section of the GLP1 molecule from degradation due to other proteases in the blood.
  • Because the rate of release of the GLP-1 analogue can be controlled by modifying the Factor Xa digestion through the modification of the Factor Xa sequence, a more constant GLP-1 analogue blood level may be achieved and thereby reduce undesirable side effects associated with varying blood sugar levels. This is generally depicted in FIG. 2 as which shows from left to right hGLP1R ECD protected Type 1 molecule 214 shedding protection sequence 204 in a controlled in predetermined manner as digestion progresses producing a released GLP1-hFc(Trulicity) molecule that provides users with substantially constant drug concentration as shown in FIGS. 4-11 discussed below.
  • One particular way was this may be accomplished includes certain modifications to portions of the Factor Xa digestion sequence (e.g., RKRR, RGER, RKR, RR etc.). Further, a point mutation (R108G) or deletion (position R108 to E116) may also be made in human GLP1R ECD in if desired to remove an internal Factor Xa cleavage site.
  • Similarly, as shown in FIG. 3 , a more particularized version of the Type 2 molecule of FIG. 1 is depicted. As shown, an hGLP1 antibody protected Type 2 molecule 314 constructed in accordance with the present invention. A protection sequence 304 (e.g., hGLP1R ECD or an anti GLP1 antibody) was linked to the N-terminus of a GLP-1 agonist 302 through 3xG4S linker 310 contains a protease cleavage site (e.g., Factor Xa) to form molecule 306, then molecule 306 was linked to the N-terminus of the light chain of an anti-GLP1R targeting antibody to form fusion protein 314. In such a fusion protein, the GLP1R ECD binds to GLP1, which, as above in FIG. 3 confers two primary benefits: 1) substantially reduces or eliminates the DPP4 cleavage, and 2) protects the middle section of the GLP1 molecule from degradation due to other proteases in the blood.
  • Because the rate of release of the GLP-1 analogue can be controlled by modifying the Factor Xa digestion through the modification of Factor Xa sequence, a more constant GLP-1 analogue blood level may be achieved and thereby reduce undesirable side effects associated with varying blood drug concentration levels. This is generally depicted in FIG. 3 which shows from left to right the Type 2 molecule 314 shedding protection sequence 304 in a controlled in predetermined manner as digestion progresses producing a released GLP1-antibody molecule 318 that provides users with substantially constant blood drug concentration as shown in the charts below.
  • EXAMPLE 1
  • The following examples reflects experiments conducted on Aug. 1, 2019 through Dec. 31, 2019 which was performed by outsourced CRO companies.
  • Reagents used and lot numbers:
  • Eli Lilly Trulicity (Lot#02448599)
  • pTG1: hFc (lot#U6443DH140S05/P90011809)
  • pTG3: hGLP1R ECD-3xG4S-RKRR-Trulicity (lot#U5585DH140S05/P90011809)
  • p7: hGLP1R ECD(R108G)-3xG4S-RGER-Trulicity (lot#U2243DL140-4/P9EA001)
  • p8: hGLP1R ECD(R108G)-3xG4S-Trulicity (lot#U2243DL140-9/P9EA001)
  • p9: hGLP1R ECD (1-116)-3xG4S-RGER-Trulicity (lot U2243DL140-14/P9EA001)
  • p10: hGLP1R ECD(R108G)-3xG4S-RR-Trulicity (lot#U2243DL140-19/P9EA001)
  • p11: hGLP1R ECD(R108G)-3xG4S-RKR-Trulicity (lot#U2243DL14024/P9EA001)
  • Experimental Procedure (include description of relevant cell lines, proteins, reagents, and instrument type and model):
  • A hGLP1R ECD molecule (SEQ ID NO 3) was linked to the N-terminus of Trulicity through a linker contains a 3xG4S and a protease cleavage site (Factor Xa, NEB lot#P8010S).
  • To better control releasing time of active Trulicity, several modifications were made for the Factor Xa cleavage sequence (e.g., RGER, RKR, RR etc.). A point mutation(R108G) (SEQ ID NO 4) or deletion (position R108 to E116) (SEQ ID NO 5) were made in human GLP1R ECD in order to remove a Factor Xa cleavage site.
  • For cAMP bioassay, the HEK293-CNG-HuGLP1R stable cells were seeded into 96-well assay plates at 70,000 cells/well in OPTIMEM supplemented with 10% FBS and then incubated at 37° C. in 5% CO2 overnight. The following day, 50 uM phosphodiesterase inhibitor R020-1724 in 1xMembrane Potential Dye (Creative Biogene, lot#FMD10) was added and incubated at 37° C. for 2 hours, and then read with fluorescence microplate reader (F0, Excitation/Emission=530nm/570nm) (MD Paradigm).
  • To determine the dose response of the test proteins, substantially all proteins were predigested with Factor Xa to release active Trulicity. Next, Trulicity, and released pTG3, p7, p8, p9, p10 and p11 (SEQ IDs 6-11 respectively) were added to cells at concentrations ranging from 0.001nM to 100nM, incubate at 37° C. for 25 minutes before read with fluorescence microplate reader (F25). The results were analyzed using nonlinear regression (three parameter) with Prism 6 software (GraphPad) to obtain EC50 values.
  • For ELISA, the plate was coated with Mouse anti-human IgG (Fc) (CELLWAYLAB lot#C010202) at 4 ug/ml, 4° C. overnight, block with 0.2% BSA/0.1%PC/0.1% at room temperate for one hour, then samples prepared from mouse serum were added and incubated at room temperate for an hour. The Trulicity is used as the standard curve. Anti GLP-1 antibody (ThermoFisher, Catalog#: ABS03310B005) is used for detecting active Trulicity. Next, after adding TMB for 10 minutes, the reaction was stopped by 2M H2SO4, and read at 450nm with plate reader (CMax Plus, Molecular Devices).
  • Pharmacokinetic profiles of p9 and pll (SEQ IDs 9 and 11 respectively) in C57/BL Mice (Beijing Vital River Laboratory Animal Technology Company) were studied after subcutaneous (SC) administration of 200 nmol/kg drugs. Blood is drawn from the test subjects (4 mice per group) at various time at 6h, day 1, 2, 4 and 7 after dosing. Serum is collected from each sample and analyzed by N-terminal specific ELISA which only detect active Trulicity. The blood drug concentration at each time point are shown in FIG. 6 .
  • The effect of p9 and pll on glucose tolerance was determined in both C57/BL and BKS/DB mouse (JiangSu GemPharmatech Company). Each group (4 mice per group) received a single subcutaneous injection of hFc control pTG1, p9, p11, and Trulicity at 200 nmol/kg. Intraperitoneal glucose tolerance tests (IPGTT) were performed on Day 3, Day 6 and 14 after overnight fasting with blood glucose measurements at 0, 15, 30, 60, and 120 minutes. Mean±SEM of blood glucose levels at each time point and glucose area under curve (AUC) were calculated for each group and shown in FIGS. 7-11 . Excel t-test was used to assess the significance to the control group, *: p<0.05, **: p<0.01, ***: p<0.001, ****: p<0.0001
  • Invitro Results Summary and Conclusions
  • As shown in the Table 1 and FIG. 4 , Trulicity™, and the released p7, p9 and p11 show EC50 values of 22 nM, 25 nM, 17.6 nM and 26.9 nM, respectively, for GLP1R activation. The EC50 for released pTG3 and p10 is only 617 nM and 120 nM, indicating the Factor Xa digestion were not at predicted sites. This was confirmed by N-terminal amino acids sequence analysis. The EC50 for p8 is undetectable because there is no Factor Xa digestion site, therefore the active Trulicity cannot be released.
  • As show in FIG. 5 , the p7, p9, p10 and p11 only show activities after Factor Xa digestion (released). The p8 (SEQ ID8) negative control shows no cAMP activity because it contains no Factor Xa digestion site and the active Trulicity couldn't be released.
  • TABLE 1
    EC50 for released GLP1 Fusion Proteins
    GLP1 fusion proteins EC50
    hGLPIR ECD-3xG4S-RKRR-Trulicity(pTG3, 617 nM
    lot#U5585DH140S05/P90011809)
    hGLPIR ECD(R108G)-3xG4S-RGER-Trulicity(p7,  25 nM
    lot#U2243DL140-4/P9EA001)
    hGLPIR ECD(R108G)-3xG4S Trulicity(p8, lot# undetectable
    U2243DL140-9/P9E A001)
    hGLPIR ECD(1-116)-3xG4S-RGER-Trulicity(p9, lot 17.6 nM 
    U2243DL140-14/P9E A001)
    hGLPIR ECD(R108G)-3xG4S-RR-Trulicity(p10, lot# 120 nM
    U2243DL140-19/P9E A001)
    hGLPIR ECD(R108G)-3xG4S-RKR-Trulicity(p11, lot# 26.9 nM 
    U2243DL140-24/P9E A001)
    Eli Lilly’s Trulicity(Lot#)  22 nM
  • Invivo Results Summary and Conclusions
  • FIG. 6 shows Pharmacokinetic profiles of released p9 and pll in C57/BL Mice after subcutaneous (SC) administration of 200 nmol/kg. The half-life for released p11 is 6 days, the half-life for p9 is longer than 7 days because 75% of released p9 is still detected at day 7. Both proteins show relative constant blood drug concentration from day 1 to day 7.
  • FIGS. 7-10 show favorable IPGTT results in C57/BL mice, that is, a single administration of p11 variant of the present invention provided significant glucose reductions at day 2 and 6.
  • Similarly, FIGS. 11 a, 11 b, and 11 c show the IPGTT results in diabetic BKS/DB mice, that is, a single administration of p9 and pll variants of the invention provided significant glucose reductions at day 3, day 6 and day 14.
  • Bioequivalents
  • The GLP1 receptor agonists of the present invention may include proteins having amino acid sequences that may vary from those of the described GLP1 receptor agonists, but that retain the ability to bind GLP1 receptor. Such variant GLP1 receptor agonists may comprise one or more additions, deletions, or substitutions of amino acids when compared to parent sequence but exhibit biological activity that is essentially equivalent to that of the described GLP1 receptor agonists. Similarly, the GLP1 receptor agonist-encoding DNA sequences of the present invention may encompass sequences that comprise one or more additions, deletions, or substitutions of nucleotides when compared to the disclosed sequence, but that encode a GLP1 receptor agonist that is essentially bioequivalent to a GLP1 receptor agonist of the invention.
  • Two proteins may be considered bioequivalent if, for example, they are pharmaceutical equivalents or pharmaceutical alternatives whose rate and extent of absorption are substantially the same do not exhibit a significant difference when administered at the same molar dose under similar experimental conditions, which may include either single dose or multiple doses. Some proteins will be considered equivalents or pharmaceutical alternatives if they are equivalent in the extent of their absorption but not in their rate of absorption and yet may be considered bioequivalent because such differences in the rate of absorption are intentional and are reflected in the labeling, are not essential to the attainment of effective body drug concentrations on, e.g., chronic use, and are considered medically insignificant for the particular drug product studied.
  • In one embodiment, two GLP1 receptor agonist proteins are bioequivalent if there are no clinically meaningful differences in their safety, purity, potency or efficacy.
  • In one embodiment, two GLP1 receptor agonist proteins are bioequivalent if a patient can be switched one or more times between the reference product and the biological product without an expected increase in the risk of adverse effects, including a clinically significant change in immunogenicity, or diminished effectiveness, as compared to continued therapy without such switching.
  • In one embodiment, two GLP1 receptor agonist proteins are bioequivalent if they both act by a common mechanism or mechanisms of action for the condition or conditions of use, to the extent that such mechanisms are known.
  • Bioequivalence may be demonstrated by in vivo and/or in vitro methods. Bioequivalence measures include, e.g., (a) an in vivo test in humans or other mammals, in which the concentration of the protein or its metabolites is measured in blood, plasma, serum, or other biological fluid as a function of time; (b) an in vitro test that has been correlated with and is reasonably predictive of human in vivo bioavailability data; (c) an in vivo test in humans or other mammals in which the appropriate acute pharmacological effect of the protein (or its target) is measured as a function of time; and (d) in a well-controlled clinical trial that establishes safety, efficacy, or bioavailability or bioequivalence of an antigen-binding protein.
  • Bioequivalent variants of the GLP1 receptor agonist proteins of the invention may be constructed by, for example, making various substitutions of residues or sequences or deleting terminal or internal residues or sequences not needed for biological activity. For example, cysteine residues not essential for biological activity can be deleted or replaced with other amino acids to prevent formation of unnecessary or incorrect intramolecular disulfide bridges upon renaturation. In other contexts, bioequivalent proteins may include variants comprising amino acid changes, which modify the glycosylation characteristics of the proteins, e.g., mutations that eliminate or remove glycosylation.
  • Therapeutic Administration and Formulations
  • The invention provides therapeutic compositions comprising the GLP1 receptor agonists. Therapeutic compositions in accordance with the invention will be administered with suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like. A multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. These formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTIN™), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. “Compendium of excipients for parenteral formulations” PDA (1998) J Pharm Sci Technol 52:238-311.
  • The dose of GLP1 receptor agonist may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like. When an antigen-binding protein of the present invention is used for treating a disease or disorder in an adult patient, or for preventing such a disease, it is advantageous to administer the antigen-binding protein of the present invention normally at a single dose of about 0.001 to about 100 mg/kg body weight, more preferably about 0.001 to about 60, about 0.01 to about 10, or about 0.01 to about 1 mg/kg body weight. Depending on the severity of the condition, the frequency and the duration of the treatment can be adjusted. In certain embodiments, the antigen-binding protein or antigen-binding fragment thereof of the invention can be administered as an initial dose of at least about 0.001 mg to about 100 mg, about 0.001 to about 50 mg, about 0.005 to about 50 mg, about 0.01 to about 40 mg, to about 30 mg, or to about 10 mg. In certain embodiments, the initial dose may be followed by administration of a second or a plurality of subsequent doses of the GLP1 receptor agonist in an amount that can be approximately the same or less than that of the initial dose, wherein the subsequent doses are separated by at least 1 day to 3 days; at least one week, at least 2 weeks; at least 3 weeks; at least 4 weeks; at least 5 weeks; at least 6 weeks; at least 7 weeks; at least 8 weeks; at least 9 weeks; at least 10 weeks; at least 12 weeks; or at least 14 weeks.
  • Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al. (1987) J. Biol. Chem. 262:4429-4432). Methods of introduction include, but are not limited to, intradermal, transdermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural and oral routes. The composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. The pharmaceutical composition can be also delivered in a vesicle, in particular a liposome (see, for example, Langer (1990) Science 249:1527-1533).
  • The use of nanoparticles to deliver the GLP1 receptor agonists of the present invention is also contemplated herein. Protein-conjugated nanoparticles may be used both for therapeutic and diagnostic applications. Nanoparticles may be developed and conjugated to antigen-binding proteins contained in pharmaceutical compositions to target cells. Nanoparticles for drug delivery have also been described in, for example, U.S. Pat. No. 8,257,740, or U.S. Pat. No. 8,246,995, each incorporated herein in its entirety.
  • If desired, the pharmaceutical compositions or treatments of the present invention may be delivered in a controlled release system. In one embodiment, a pump may be used. In other embodiments, polymeric materials can be used. In yet other embodiments, a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose.
  • The injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous, intracranial, intraperitoneal and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antigen-binding protein or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections. As the aqueous medium for injections, there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc. As the oily medium, there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc. The injection thus prepared is preferably filled in an appropriate ampoule.
  • A pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe. In addition, with respect to subcutaneous delivery, a pen or wand type delivery device readily has applications in delivering a pharmaceutical composition of the present invention. Such a pen delivery device can be reusable or disposable. A reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused. In a disposable pen delivery device, there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention. Examples include, but certainly are not limited to AUTOPEN™ (Owen Mumford, Inc., Woodstock, UK), DISETRONIC™ pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25™ pen, HUMALOG™ pen, HUMALIN 70/30™ pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPEN™ I, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIOR™ (Novo Nordisk, Copenhagen, Denmark), BD™ pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPEN™, OPTIPEN PRO™, OPTIPEN STARLET™, and OPTICLIK™ (Sanofi-Aventis, Frankfurt, Germany), to name only a few. Examples of disposable pen delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but certainly are not limited to the SOLOSTAR™ pen (Sanofi-Aventis), the FLEXPEN™ (Novo Nordisk), and the KWIKPEN™ (Eli Lilly), the SURECLICK™ Autoinjector (Amgen, Thousand Oaks, Calif.), the PENLET™ (Haselmeier, Stuttgart, Germany), the EPIPEN (Dey, L.P.) and the HUMIRA™ Pen (Abbott Labs, Abbott Park, Ill.), to name only a few.
  • Advantageously, the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients. Such dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc. The amount of the GLP1 receptor agonist contained is generally about 0.001 to about 100 mg per dosage form in a unit dose; especially in the form of injection, it is preferred that the GLP1 receptor agonist is contained in about 0.001 to about 100 mg and in about 0.01 to about 100 mg for the other dosage forms.
  • Therapeutic Uses of the GLP1 Receptor Agonists
  • The GLP1 receptor agonists of the present invention are useful for the treatment, and/or prevention of certain adverse medical conditions associated with hyperglycemia including diabetes. It also may be useful for lessening the severity of at least one symptom associated with such conditions. This may include, for example administering it at a therapeutic dose to a patient with diabetes (e.g., type 2 diabetes).
  • Furthermore, in some embodiments, the GLP1 receptor agonists of the invention are useful to treat subjects suffering from health problems form the group comprising diabetes mellitus, obesity, insulin resistance, hypertension, dyslipidemia, Type 2 diabetes, Type 1 diabetes, pre-diabetes, cardiovascular disease, atherosclerosis, congestive heart failure, coronary heart disease, arteriosclerosis, peripheral artery disease, stroke, respiratory dysfunction, renal disease, fatty liver disease, metabolic syndrome and similar or associated conditions.
  • In some embodiments, the GLP1 receptor agonists of the invention are useful to treat subjects that are overweight, obese and/or prevent or treat one or more obesity-associated disorders such as heart disease, stroke, and diabetes.
  • In some embodiments, the GLP1 receptor agonists of the invention are useful to treat subjects suffering from diabetes and/or prevent one or more complications of associated with diabetes such as heart disease, stroke, kidney disease, retinopathy, blindness and peripheral nerve damage.
  • It will be understood that one or more GLP1 receptor agonist fusion proteins of the present invention may be used as a preventive measure for patients in danger of developing diabetes (e.g., from type 2 diabetes). Such risks include, but are not limited to, patients of advanced age, pregnant women, and/or other risk factors including family history of obesity, high blood cholesterol, smoking, excessive alcohol consumption, and/or lack of exercise.
  • In a further embodiment, the proteins of the invention may be used for the preparation of a pharmaceutical composition or medicament for treating patients suffering from a disease or disorder such as diabetes and obesity. In other embodiments of the invention, the GLP1 receptor agonists may be used as complimentary or supplemental therapy with any other suitable therapy known to those skilled in the art useful for treating or ameliorating a disease or disorder associated with hyperglycemia such as diabetes (e.g., type 2 diabetes).
  • Combination Therapies
  • Combination therapies contemplated by the present invention may include a GLP1 receptor agonist of the invention and any suitable additional therapeutic agent that may be advantageously combined therewith, such as GLP1 receptor agonist of the invention, or a biologically active fragment of the invention as would be appreciated by one of ordinary skill in the art. Further combination may include the GLP1 receptor agonists of the present inventio combined synergistically with one or more drugs or therapy used to treat any disease or disorder associated with hyperglycemia (e.g., diabetes). In some embodiments, the GLP1 receptor agonists of the invention may be combined with one or more other therapeutic agent(s) to reduce blood sugar levels in a subject, or to ameliorate one or more symptoms of diabetes.
  • The GLP1 receptor agonists of the present invention may be used in combination with an insulin (insulin or an insulin analog), insulin sensitizers such as biguanides (e.g., metformin), and thiazolidinediones (e.g., rosiglitazone), insulin secretagogues such as sulphonylureas (e.g., chlorpropamide), and glinides (e.g., nateglinide), alpha-glucosidase inhibitors (e.g., acarbose), dipeptidyl peptidase 4 (DPP4) inhibitors (e.g., sitagliptin), pramlinitide, bromocriptine, sodium glucose cotransporter 2 (SGLT-2) inhibitors (e.g., canagliflozin), an anti-hypertensive drug (e.g., an angiotensin-converting enzyme inhibitor, an angiotensin receptor blocker, a diuretic, a calcium channel blocker, an alpha-adrenoceptor blocker, an endothelin-1 receptor blocker, an organic nitrate, and a protein kinase C inhibitor), a statin, aspirin, a different GLP1 receptor agonist, a dietary supplement or any other therapy (e.g., exercise) to treat or manage diabetes.
  • In yet other embodiments, the GLP1 receptor agonists may be administered in combination with a one or more therapeutic agent(s) including: insulin, an insulin analog, metformin, rosiglitazone, pioglitazone, chlorpropamide, glibenclamide, glimepiride, glipizide, tolazamide, tolbutamide, nateglinide, repaglinide, acarbose, miglitol, exenatide, liraglutide, albiglutide, dulaglutide, sitagliptin, saxagliptin, linagliptin, alogliptin, pramlinitide, bromocriptine quick-release, canagliflozin, dapagliflozin, empagliflozin, diet modifications and exercise.
  • As used herein, the term “in combination with” means that additional therapeutically active component(s) may be administered prior to, concurrent with, or after the administration of the GLP1 receptor agonist of the present invention. The term “in combination with” also includes sequential or concomitant administration of a GLP1 receptor agonist and a second therapeutic agent.
  • The additional therapeutically active component(s) may be administered to a subject prior to administration of a GLP1 receptor agonist of the present invention. For example, a first component may be deemed to be administered “prior to” a second component if the first component is administered 1 week before, 72 hours before, 60 hours before, 48 hours before, 36 hours before, 24 hours before, 12 hours before, 6 hours before, 5 hours before, 4 hours before, 3 hours before, 2 hours before, 1 hour before, 30 minutes before, 15 minutes before, 10 minutes before, 5 minutes before, or less than 1 minute before administration of the second component. In other embodiments, the additional therapeutically active component(s) may be administered to a subject after administration of a GLP1 receptor agonist of the present invention. For example, a first component may be deemed to be administered “after” a second component if the first component is administered 1 minute after, 5 minutes after, 10 minutes after, 15 minutes after, 30 minutes after, 1 hour after, 2 hours after, 3 hours after, 4 hours after, 5 hours after, 6 hours after, 12 hours after, 24 hours after, 36 hours after, 48 hours after, 60 hours after, 72 hours after administration of the second component. In yet other embodiments, the additional therapeutically active component(s) may be administered to a subject concurrent with administration of a GLP1 receptor agonist of the present invention. “Concurrent” administration, for purposes of the present invention, includes, e.g., administration of a GLP1 receptor agonist and an additional therapeutically active component to a subject in a single dosage form, or in separate dosage forms administered to the subject within about 30 minutes or less of each other. If administered in separate dosage forms, each dosage form may be administered via the same route (e.g., both the GLP1 receptor agonist and the additional therapeutically active component may be administered intravenously, etc.); alternatively, each dosage form may be administered via a different route (e.g., the GLP1 receptor agonist may be administered intravenously, and the additional therapeutically active component may be administered orally). In any event, administering the components in a single dosage from, in separate dosage forms by the same route, or in separate dosage forms by different routes are all considered “concurrent administration,” for purposes of the present disclosure. For purposes of the present disclosure, administration of a GLP1 receptor agonist “prior to”, “concurrent with,” or “after” (as those terms are defined herein above) administration of an additional therapeutically active component is considered administration of a GLP1 receptor agonist “in combination with” an additional therapeutically active component.
  • The present invention includes pharmaceutical compositions in which a GLP1 receptor agonist of the present invention is co-formulated with one or more of the additional therapeutically active component(s) as described elsewhere herein.
  • All publications and patent documents disclosed or referred to herein are incorporated by reference in their entirety. The foregoing description has been presented only for purposes of illustration and description. This description is not intended to limit the invention to the precise form disclosed. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figures. Such modifications are intended to fall within the scope of the appended claims.
  • It will be understood that the molecular structures and methods disclosed herein are merely illustrative and are not meant to be comprehensive or necessarily performed in the order or exact fashion shown. Persons skilled in the art will appreciate that the present invention can be practiced by other than the described embodiments, which are presented for purposes of illustration rather than of limitation, and the present invention is limited only by the claims which follow.
  • The below Sequence ID is also submitted in an ASCII text file named “YW-001_SEQ” and is specifically incorporated herein by reference.

Claims (20)

1. A fusion protein comprising:
a protection sequence for Glucagon-like peptide (GLP-1) to impede in-vivo cleavage, inactivation or degradation;
an amino acid linker region containing a protease cleavage site configured for coupling the protection sequence to a GLP-1 analogue.
2. The fusion protein of claim 1, wherein the protection sequence further comprises a Glucagon-like peptide 1 (GLP-1) Receptor extracellular domain (ECD).
3. The fusion protein of claim 2, wherein the Glucagon-like peptide 1 Receptor ECD includes a point mutation to eliminate Factor Xa cleavage site.
4. The fusion protein of claim 2, wherein the Glucagon-like peptide 1 Receptor ECD contains a deletion to remove Factor Xa cleavage site.
5. The fusion protein of claim 2, is fused to a GLP-1 analogue, preferably the N-terminus of Trulicity through a linker.
6. The fusion protein of claim 5, wherein the linker comprises a 1xG4S(GGGGS), 2xG4S(GGGGSGGGGS), or 3XG4S(GGGGSGGGGSGGGGS) followed by a protease cleavage sequence.
7. The fusion protein of claim 6, wherein the preferred protease sequence is for Factor Xa and its variant from the group comprising RKRR, RGER, RKR, RR.
8. The fusion protein of claim 5, wherein the Trulicity's Fc part is replaced by one of an anti Glucagon-like peptide 1 Receptor targeting antibody and a bispecific antibody
9. The fusion protein of claim 8, wherein the bispecific antibody is targeting to any of the two receptors form glucagon/secretin superfamily (e.g., GLP1R and GIPR).
10. The fusion protein of claim 1, wherein the protection sequence comprises an anti GLP1 antibody (an antibody's Fab region, a nanobody or bispecific T-cell engager “BiTE” antibody in certain preferred embodiments) which binds to GLP1 and protect it from protease cleavage.
11. The fusion protein of claim 1, wherein the GLP1 peptide could be other similar peptides or their hybrids in Glucagon/secretin superfamily.
12. The fusion protein of claim 11, wherein the Glucagon/secretin superfamily includes at least one of Glucagon, GLP-1, GLP-2, GIP, VIP, PACAP, Exendin-4, GLP1/GIP dual agonist and GLP1/GIP/Glucagon triple agonist.
13. A pharmaceutical comprising a fusion protein of claim 1 and a pharmaceutically acceptable carrier or diluent.
14. A method of lowering blood sugar level comprising administering a pharmaceutical composition comprising a therapeutically effective amount of the protein of claim 1 to a subject in need thereof
15. The method of claim 14, wherein the subject has a disease or disorder selected from the group consisting of diabetes mellitus, obesity, insulin resistance, hypertension, dyslipidemia, Type 2 diabetes, Type 1 diabetes, prediabetes, cardiovascular disease, atherosclerosis, congestive heart failure, coronary heart disease, arteriosclerosis, peripheral artery disease, stroke, respiratory dysfunction, renal disease, fatty liver disease, non - alcoholic steatohepatitis (NASH), and metabolic syndrome.
16. A method of preventing, treating or ameliorating at least one symptom, indication or complication of Type 2 diabetes, the method comprising administering a pharmaceutical composition comprising a therapeutically effective amount of the protein of claim 1 to a subject in need thereof
17. The method of claim 16, wherein the at least one symptom, indication or complication is selected from the group consisting of high blood sugar level, excessive thirst, increased urination, presence of ketones in urine, fatigue, weight fluctuations, blurred vision, slow healing sores, frequent infections, swollen or tender gums, obesity, heart disease, stroke, kidney disease, eye disease, nerve damage and high blood pressure.
18. The method of claims 14, wherein the pharmaceutical composition is administered in combination with a second therapeutic agent or therapy.
19. The method of claim 18, wherein the second therapeutic agent or therapy is selected from the group consisting of an insulin or insulin analogue, metformin, a thiazolidinedione, a sulfonylurea, a biguanide, chlorpropamide, a glinide, an alpha glucosidase inhibitor, nateglinide, a DPP4 inhibitor, pramlintide, sitagliptin, bromocriptine, a SGLT2 inhibitor, canagliflozin, an antihypertensive drug, a statin, aspirin, dietary modification, exercise, and a dietary supplement.
20. The method of claims 14, wherein the pharmaceutical composition is administered subcutaneously, intravenously, intradermally, intraperitoneally, orally, or intramuscularly.
US17/184,248 2020-09-22 2021-02-24 Long-lasting glp1 analogue drug for type-2 diabetes Pending US20220389071A9 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US17/184,248 US20220389071A9 (en) 2020-09-22 2021-02-24 Long-lasting glp1 analogue drug for type-2 diabetes
EP21873103.2A EP4216987A1 (en) 2020-09-22 2021-03-15 A long-lasting glp1 analogue drug for type-2 diabetes
PCT/US2021/022296 WO2022066212A1 (en) 2020-09-22 2021-03-15 A long-lasting glp1 analogue drug for type-2 diabetes
CN202180001518.5A CN114980916A (en) 2020-09-22 2021-03-15 Long-acting GLP1 analog drugs for type 2 diabetes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063081363P 2020-09-22 2020-09-22
US17/184,248 US20220389071A9 (en) 2020-09-22 2021-02-24 Long-lasting glp1 analogue drug for type-2 diabetes

Publications (2)

Publication Number Publication Date
US20220267402A1 US20220267402A1 (en) 2022-08-25
US20220389071A9 true US20220389071A9 (en) 2022-12-08

Family

ID=80845725

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/184,248 Pending US20220389071A9 (en) 2020-09-22 2021-02-24 Long-lasting glp1 analogue drug for type-2 diabetes

Country Status (4)

Country Link
US (1) US20220389071A9 (en)
EP (1) EP4216987A1 (en)
CN (1) CN114980916A (en)
WO (1) WO2022066212A1 (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE444741T1 (en) * 2006-05-10 2009-10-15 Biocompatibles Uk Ltd Spherical microcapsules containing GLP-1 peptides, their production and their use
EP2414517B1 (en) * 2009-03-30 2016-09-21 Portola Pharmaceuticals, Inc. Antidotes for factor xa inhibitors and methods of using the same
CN104302772B (en) * 2012-05-18 2017-11-10 爱德迪安(北京)生物技术有限公司 Albumen, protein conjugate and its application for treating diabetes
AU2017235783A1 (en) * 2016-03-15 2018-09-20 Cancer Research Technology Limited Antibodies and related molecules and uses thereof
EP3829622A4 (en) * 2018-08-02 2022-05-11 Duke University Dual agonist fusion proteins
CN110964116A (en) * 2018-09-26 2020-04-07 北京辅仁瑞辉生物医药研究院有限公司 GLP1-Fc fusion proteins and conjugates thereof

Also Published As

Publication number Publication date
US20220267402A1 (en) 2022-08-25
CN114980916A (en) 2022-08-30
EP4216987A1 (en) 2023-08-02
WO2022066212A1 (en) 2022-03-31

Similar Documents

Publication Publication Date Title
US11253574B2 (en) Fusion proteins and methods of use
US11779633B2 (en) Glucagon-like peptide 1 receptor agonists and uses thereof
KR20210024583A (en) How to use GIP/GLP1 coagonists for therapy
AU2022202506A1 (en) Treatment of post-bariatric hypoglycemia with GLP-1 antagonists
US20220389071A9 (en) Long-lasting glp1 analogue drug for type-2 diabetes
EA045412B1 (en) GLUCAGON-LIKE PEPTIDE 1 RECEPTOR AGONISTS AND THEIR APPLICATIONS
Cases Glucagon-like peptide 1 (GLP-1) receptor agonists in the management of the patient with type 2diabetes mellitus and chronic kidney disease: an approach for the nephrologist
Frias et al. To assess the efficacy and safety of the glucagon-like peptide 1 receptor agonist (GLP-1 RA) efpeglenatide versus placebo in patients with type 2 diabetes inade-quately controlled with diet and exercise alone.
WO2022132712A1 (en) Methods of treating diabetes
WO2024064842A1 (en) Methods of treating obesity, diabetes, and liver dysfunction
Lorraine Lipscombe et al. Pharmacologic Glycemic Management of Type 2 Diabetes in Adults: 2020 Update

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: SERPENTIDE INC., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WEI, YANG;REEL/FRAME:062395/0627

Effective date: 20221212

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED