US20160039877A1 - Compositions and methods of using islet neogenesis peptides and analogs thereof - Google Patents

Compositions and methods of using islet neogenesis peptides and analogs thereof Download PDF

Info

Publication number
US20160039877A1
US20160039877A1 US14/775,568 US201314775568A US2016039877A1 US 20160039877 A1 US20160039877 A1 US 20160039877A1 US 201314775568 A US201314775568 A US 201314775568A US 2016039877 A1 US2016039877 A1 US 2016039877A1
Authority
US
United States
Prior art keywords
seq
peptide
analog
peptides
analogs
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/775,568
Other languages
English (en)
Inventor
Liping Liu
Ru Bai
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.)
Shenzhen Hightide Biopharmaceutical Ltd
Original Assignee
Shenzhen Hightide Biopharmaceutical Ltd
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 Shenzhen Hightide Biopharmaceutical Ltd filed Critical Shenzhen Hightide Biopharmaceutical Ltd
Assigned to SHENZHEN HIGHTIDE BIOPHARMACEUTICAL, LTD. reassignment SHENZHEN HIGHTIDE BIOPHARMACEUTICAL, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAI, Ru, LIU, LIPING
Publication of US20160039877A1 publication Critical patent/US20160039877A1/en
Assigned to SHENZHEN HIGHTIDE BIOPHARMACEUTICAL, LTD. reassignment SHENZHEN HIGHTIDE BIOPHARMACEUTICAL, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAI, Ru, LIU, LIPING
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4705Regulators; Modulating activity stimulating, promoting or activating activity
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4733Acute pancreatitis-associated protein
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4738Cell cycle regulated proteins, e.g. cyclin, CDC, INK-CCR
    • 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
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/02Linear peptides containing at least one abnormal peptide link
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates generally to the field of medicine and pharmaceuticals, and more specifically to peptide therapies for treating diabetes and other diseases.
  • T1D type 1 DM
  • T2D type 2 DM
  • T1D results from the body's failure to produce insulin, and requires the patient to administer insulin daily.
  • T2D results from insulin resistance, a condition in which cells fail to use insulin properly.
  • non-insulin therapies for T2D.
  • T2D there is a large portion of late stage T2D patients requiring insulin administration due to the loss of ⁇ -cell function as the disease progresses.
  • pancreatic islet mass At the time of diagnosis, over 90% of islet mass has been lost in T1D patients, and approximately 50% has been lost in T2D patients. Many attempts have been made in quest of a potential stimulus for islet neogenesis, which is considered as the optimal treatment for both T1D and T2D.
  • INGAP islet neogenesis-associated protein
  • HIP human proIslet peptide
  • GLP-1 glucagon like peptide-1
  • VIP islet endocrine neuropeptide vasoactive intestinal peptide
  • epidermal growth factor and gastrin are capable of inducing pancreatic progenitor cells, located in the nonendocrine fraction of the pancreas, to differentiate into fully functional islets in various animal models.
  • INGAP peptide a 15-mer peptide derived from the sequence of INGAP at amino acids 104-118, has been shown to induce islet neogenesis in multiple animal models, reverse streptozotocin (STZ) induced diabetes in mice, increase C-peptide secretion in T1D patients, and improve glycemic control in T2D patients. Additional biological effects of INGAP-PP have been reported, including dose dependent stimulation of expansion of ⁇ -cell mass and increased insulin secretion and ⁇ -cell size.
  • HIP the bioactive peptide encoded by a portion of the human REG3A gene
  • HIP the human homolog of the INGAP peptide.
  • Previous studies have shown that treatment of human pancreatic ductal tissues with HIP stimulated the production of insulin.
  • Administration of HIP improved glycemic control and increased islet number in diabetic mice.
  • the stabilized form of HIP is currently being tested in a single ascending dose clinical trial with the goal of exploring the tolerability, safety and pharmacokinetics. It is of note that total daily doses of 60, 120, 240, 480, and 720 mg are planned. Although there is a lack of data on the efficacious dose of HIP, the planned clinical trial doses infer the peptide's low potency.
  • the invention provides peptides and analogs of INGAP and HIP peptides.
  • the peptides and analogs can be used in methods for treating various disease and conditions.
  • diseases and conditions can include impaired pancreatic function, treating a metabolic disease, for example, diabetes, both type 1 and type 2 diabetes, islets induction, expansion and proliferation for transplantation, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation.
  • FIG. 1 shows the comparison of ARIP cell (a rat pancreatic ductal cell line) proliferation in the presence of 100 nM of INGAP Scrambled PP 1 (Peptide 3), INGAP-PP (Peptide 1), and a selected peptide analog, Peptide 7 (see Table 2).
  • FIG. 2 shows a stability comparison in culture medium of INGAP-PP (Peptide 1) and selected peptide analogs, Peptide 7 and Peptide 8 (see Table 2).
  • FIG. 3 shows a stability comparison in mouse plasma of INGAP-PP (Peptide 1) and selected peptide analogs, Peptide 12, Peptide 16 and Peptide 29 (see Table 2).
  • FIG. 4 shows a stability comparison in human plasma of INGAP-PP (Peptide 1) and selected peptide analogs, Peptide 12 and Peptide 16 (see Table 2).
  • FIG. 5 shows a stability comparison in mouse plasma of HIP (Peptide 2) and selected peptide analogs, Peptide 52 and Peptide 54 (see Table 3).
  • FIGS. 6A-6C show the efficacy comparison of INGAP-PP (Peptide 1), INGAP Scrambled PP 1 (Peptide 3) and a selected peptide analog, Peptide 7 (see Table 2) in STZ induced diabetic mice model.
  • FIG. 6A Blood glucose (BG) after 21 day treatment
  • FIG. 6B Fasting insulin levels after 21 day treatment
  • FIG. 6C Area under curve (AUC) of glucose measured in oral glucose tolerance test (OGTT) after 21 day treatment.
  • BG Blood glucose
  • OGTT oral glucose tolerance test
  • FIG. 7 shows the number of islets defined by area ranges (arbitrary morphometric units) for equal randomly selected fields (n ⁇ 7) for animals treated with naive and Peptide 3, Peptide 1 or Peptide 7.
  • FIG. 8 shows the increase of glucose-stimulated insulin secretion of islets with or without the co-incubation of selected peptides (10 ⁇ g/mL), Peptide 12, Peptide 16 and Peptide 1 (see Table 2). Co-incubation with 100 nM Glucagon like peptide-1 (GLP-1) was included as a positive control.
  • the present invention provides compounds, in particular peptide or peptide analogs, that exhibit properties useful for treating a variety of diseases and conditions, particularly diseases and conditions relating to diabetes.
  • the peptides and analogs of the invention are additionally useful for treating impaired pancreatic function, treating a metabolic disease, ex vivo islet induction, expansion and proliferation for transplantation, increasing the survival of transplanted islets in vivo, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation.
  • the present invention provides a series of INGAP-PP and HIP analogs with comparable or improved stability and activities compared to the wild-type peptides (see Tables 2 and 3).
  • the improved pharmaceutical properties of these peptide analogs make them particularly suitable for clinical development.
  • the present invention also provides pharmaceutical compositions comprising a compound according to the present invention and the use of compounds according to the present invention for preparing medicaments for treating metabolic diseases, including but not limited to type 1 diabetes (T1D) and type 2 diabetes (T2D).
  • T1D type 1 diabetes
  • T2D type 2 diabetes
  • the invention further provides the compositions of the invention in suitable formulations, including sustained release formulations.
  • INGAP islet neogenesis associated protein
  • HIP human proIslet peptide
  • the present invention provides analogs of INGAP-PP and HIP peptides, including the peptides or analogs of Tables 2 and 3 that are not parent INGAP-PP or HIP peptides, that exhibit unexpected and beneficial properties over the INGAP-PP and HIP peptides.
  • peptide refers to a polymer of two or more amino acids.
  • the peptide can be modified to include analogs, derivatives, functional mimetics, pseudopeptides, and the like, so long as the peptide comprises a polymer of at least two amino acids.
  • the meaning of the term “peptide” is well known to those skilled in the art.
  • a peptide includes two or more amino acids joined by an amide bond between the carboxyl group of one amino acid residue and the amino group of the adjacent amino acid residue.
  • a peptide can comprise naturally occurring amino acids or non-naturally occurring amino acids.
  • an analog refers to a variant of a parent molecule, for example, a parent peptide.
  • an analog of a parent peptide can include a variant, where one or more amino acids are substituted relative to the parent peptide.
  • An analog can also include a modification of a parent peptide, including but not limited to, non-naturally occurring amino acids, D amino acids, modified amino- and/or carboxy-terminal (N- or C-terminal) amino acids, in particular modifications of the amino group on the N-terminus and/or modification of the carboxyl group in the C-terminus, fatty acid modifications, peptidomimetics, pseudopeptides, and the like, as disclosed herein. Exemplary modifications are described in more detail below.
  • impaired pancreatic function refers to a disease or condition associated with the pancreas, where the pancreas exhibits a decreased function compared to that of a normal or healthy individual.
  • diseases or conditions associated with impaired pancreatic function include, but are not limited to, type 1 diabetes, type 2 diabetes, latent autoimmune diabetes in adults (LADA), impaired fasting glucose, impaired glucose tolerance, insulin deficiency, fasting hyperinsulinemia, insulin resistance, or impaired fasting insulin levels, or a combination thereof. Such diseases and conditions are discussed in more detail below.
  • the invention provides peptide analogs of INGAP-PP and HIP peptides.
  • Table 1 shows the sequence of INGAP-PP and HIP peptides, as well as various scrambled versions of the peptides that are used as negative controls in experiments described herein or can be used as negative controls in comparative studies with INGAP-PP, HIP or the peptides analogs of the invention.
  • INGAP-PP various analogs of INGAP-PP are provided as peptides or analogs of the invention.
  • Exemplary INGAP-PP peptide analogs of the invention are provided in Table 2.
  • HIP HIP peptides or analogs of the invention.
  • exemplary HIP peptide analogs of the invention are provided in Table 3.
  • the invention provides peptides or analogs thereof that are analogs of INGAP-PP.
  • the invention provides a peptide or analog thereof comprising a sequence selected from the group consisting of IGLHDPSHGTLPAGS (SEQ ID NO:7); and IGLHDPSHGTLPAG (SEQ ID NO:73).
  • the peptide or analog can comprise a peptide or analog selected from: IGLHDPSHGTLPAGS (SEQ ID NO:7); IGLHDPSHGTLPAG (SEQ ID NO:73); IGLHDPSHGTLPAGSK (SEQ ID NO:9); IGLHDPSHGTLP(Aib)GS (SEQ ID NO:10); IGLHDPSHGTLP(N-methyl-L-Ala)GS (SEQ ID NO:11); Ac-IGLHDPSHGTLPAGS (SEQ ID NO:12); (D-Ile)GLHDPSHGTLPAGS (SEQ ID NO:13); (L-NorVal)GLHDPSHGTLPAGS (SEQ ID NO:14); (L-NorLeu)GLHDPSHGTLPAGS (SEQ ID NO:15); IGLHDPSHGTLPAG-NH2 (SEQ ID NO:28); Ac-IGLHDPSHGTLPAGS-NH2 (SEQ ID NO:29); Ac-IGLHDPSHGTLPAG-NH2 (SEQ
  • the peptide or analog thereof can consist of: IGLHDPSHGTLPAGS (SEQ ID NO:7); IGLHDPSHGTLPAG (SEQ ID NO:73); IGLHDPSHGTLPAGSK (SEQ ID NO:9); IGLHDPSHGTLP(Aib)GS (SEQ ID NO:10); IGLHDPSHGTLP(N-methyl-L-Ala)GS (SEQ ID NO:11); Ac-IGLHDPSHGTLPAGS (SEQ ID NO:12); (D-Ile)GLHDPSHGTLPAGS (SEQ ID NO:13); (L-NorVal)GLHDPSHGTLPAGS (SEQ ID NO:14); (L-NorLeu)GLHDPSHGTLPAGS (SEQ ID NO:15); IGLHDPSHGTLPAG-NH2 (SEQ ID NO:28); Ac-IGLHDPSHGTLPAGS-NH2 (SEQ ID NO:29); Ac-IGLHDPSHGTLPAG-NH2 (SEQ ID NO:31); Ac-
  • An embodiment of the invention provided herein includes a peptide or analog thereof comprising a peptide or analog selected from the group consisting of: Ac-IGLHDPSHGTLPNGS (SEQ ID NO:16); (D-Ile)GLHDPSHGTLPNGS (SEQ ID NO:17); Ac-IGLHD PSHGT LPNGS-NH2 (SEQ ID NO:31); IGLHDPSHGTLPNGS-NH2 (SEQ ID NO:32); IGLHDPSHGTLPNGSC (SEQ ID NO:33); Ac-IGLHDPSHGTLPNGSC (SEQ ID NO:34); IGLHDPSHGTLPNGSC-NH2 (SEQ ID NO:35); Ac-IGLHDPSHGTLPNGSC-NH2 (SEQ ID NO:36); IGLHDPSHGTLPNGC (SEQ ID NO:37); Ac-IGLHDPSHGTLPNGC (SEQ ID NO:38); IGLHDPSHGTLPNGC-NH2
  • the peptide or analog thereof consists of: Ac-IGLHDPSHGTLPNGS (SEQ ID NO:16); (D-Ile)GLHDPSHGTLPNGS (SEQ ID NO:17); Ac-IGLHD PSHGT LPNGS-NH2 (SEQ ID NO:31); IGLHDPSHGTLPNGS-NH2 (SEQ ID NO:32); IGLHDPSHGTLPNGSC (SEQ ID NO:33); Ac-IGLHDPSHGTLPNGSC (SEQ ID NO:34); IGLHDPSHGTLPNGSC-NH2 (SEQ ID NO:35); Ac-IGLHDPSHGTLPNGSC-NH2 (SEQ ID NO:36); IGLHDPSHGTLPNGC (SEQ ID NO:37); Ac-IGLHDPSHGTLPNGC (SEQ ID NO:38); IGLHDPSHGTLPNGC-NH2 (SEQ ID NO:39); Ac-IGLHDPSHGTLPNGC-NH2 (SEQ ID NO:40); IGLHDPSHGTLPNGS (SEQ
  • the invention provides a peptide or analog thereof comprising a sequence selected from the group consisting of: IGLHAPSHGTLPNGS (SEQ ID NO:6); IGLHAPSHGTLPAGS (SEQ ID NO:8); IGLHDPSHGTEPNGS (SEQ ID NO:18); IGLHDPSQGTLPNGS (SEQ ID NO:19); IGLHDPTHGTLPNGS (SEQ ID NO:20); IGLHDPSHGTLPNGE (SEQ ID NO:21); IGLHDPSHGTLPNGK (SEQ ID NO:22); IGLHDPSHGTLPAGK (SEQ ID NO:23); IGLHDPSHGTEPAGS (SEQ ID NO:24); IGLHDPSQGTLPAGS (SEQ ID NO:25); and IGLHDPTHGTLPAGS (SEQ ID NO:26); IGLHDPSHGTLPAGE (SEQ ID NO:27).
  • the invention provides a peptide or analog thereof comprising a peptide or analog selected from: IGLHAPSHGTLPNGS (SEQ ID NO:6); IGLHAPSHGTLPAGS (SEQ ID NO:8); IGLHDPSHGTEPNGS (SEQ ID NO:18); IGLHDPSQGTLPNGS (SEQ ID NO:19); IGLHDPTHGTLPNGS (SEQ ID NO:20); IGLHDPSHGTLPNGE (SEQ ID NO:21); IGLHDPSHGTLPNGK (SEQ ID NO:22); IGLHDPSHGTLPAGK (SEQ ID NO:23); IGLHDPSHGTEPAGS (SEQ ID NO:24); IGLHDPSQGTLPAGS (SEQ ID NO:25); IGLHDPTHGTLPAGS (SEQ ID NO:26); and IGLHDPSHGTLPAGE (SEQ ID NO:27).
  • IGLHAPSHGTLPNGS SEQ ID NO:6
  • IGLHAPSHGTLPAGS SEQ ID NO:8
  • the invention provides a peptide or analog thereof consisting of: IGLHAPSHGTLPNGS (SEQ ID NO:6); IGLHAPSHGTLPAGS (SEQ ID NO:8); IGLHDPSHGTEPNGS (SEQ ID NO:18); IGLHDPSQGTLPNGS (SEQ ID NO:19); IGLHDPTHGTLPNGS (SEQ ID NO:20); IGLHDPSHGTLPNGE (SEQ ID NO:21); IGLHDPSHGTLPNGK (SEQ ID NO:22); IGLHDPSHGTLPAGK (SEQ ID NO:23); IGLHDPSHGTEPAGS (SEQ ID NO:24); IGLHDPSQGTLPAGS (SEQ ID NO:25); IGLHDPTHGTLPAGS (SEQ ID NO:26); and IGLHDPSHGTLPAGE (SEQ ID NO:27).
  • the invention additionally provides HIP analogs.
  • the invention provides a peptide or analog thereof comprising the sequence IGLHDPTQGTEPAGE (SEQ ID NO:50).
  • the peptide or analog can comprise a peptide or analog selected from: IGLHDPTQGTEPAGE (SEQ ID NO:50); IGLHDPTQGTEP(Aib)GE (SEQ ID NO:51); Ac-IGLHDPTQGTEPAGE (SEQ ID NO:52); (D-Ile)GLHDPTQGTEPAGE (SEQ ID NO:53); Ac-IGLHDPTQGTEPAG-NH2 (SEQ ID NO:60); Ac-IGLHD PTQGT EPAGE-NH2 (SEQ ID NO:62); IGLHDPTQGTEPAGE-NH2 (SEQ ID NO:68); IGLHDPTQGTEPAGC (SEQ ID NO:69); Ac-IGLHDPTQGTEPAGC (SEQ ID NO:70); IGLHDPTQGTEPAGC-NH2
  • the peptide or analog thereof consists of: IGLHDPTQGTEPAGE (SEQ ID NO:50); IGLHDPTQGTEP(Aib)GE (SEQ ID NO:51); Ac-IGLHDPTQGTEPAGE (SEQ ID NO:52); (D-Ile)GLHDPTQGTEPAGE (SEQ ID NO:53); and Ac-IGLHDPTQGTEPAG-NH2 (SEQ ID NO:60); Ac-IGLHD PTQGT EPAGE-NH2 (SEQ ID NO:62); IGLHDPTQGTEPAGE-NH2 (SEQ ID NO:68); IGLHDPTQGTEPAGC (SEQ ID NO:69); Ac-IGLHD PTQGT EPAGC (SEQ ID NO:70); IGLHD PTQGT EPAGC-NH2 (SEQ ID NO:71); or Ac-IGLHD PTQGT EPAGC-NH2 (SEQ ID NO:72).
  • the invention provides additional HIP peptide analogs.
  • the invention provides a peptide or analog thereof comprising a peptide or analog selected from the group consisting of: Ac-IGLHDPTQGTEPNGE (SEQ ID NO:54); (D-Ile)GLHDPTQGTEPNGE (SEQ ID NO:55); Ac-IGLHDPTQGTEPNGE-NH2 (SEQ ID NO:61); IGLHDPTQGTEPNGE-NH2 (SEQ ID NO:63); IGLHDPTQGTEPNGC (SEQ ID NO:64); Ac-IGLHDPTQGTEPNGC (SEQ ID NO:65); IGLHDPTQGTEPNGC-NH2 (SEQ ID NO:66); and Ac-IGLHDPTQGTEPNGC-NH2 (SEQ ID NO:67).
  • the peptide or analog thereof can consist of: Ac-IGLHDPTQGTEPNGE (SEQ ID NO:54); (D-Ile)GLHDPTQGTEPNGE (SEQ ID NO:55); Ac-IGLHDPTQGTEPNGE-NH2 (SEQ ID NO:61); IGLHDPTQGTEPNGE-NH2 (SEQ ID NO:63); IGLHDPTQGTEPNGC (SEQ ID NO:64); Ac-IGLHDPTQGTEPNGC (SEQ ID NO:65); IGLHDPTQGTEPNGC-NH2 (SEQ ID NO:66); or Ac-IGLHDPTQGTEPNGC-NH2 (SEQ ID NO:67).
  • a peptide or analog thereof can comprise a sequence selected from the group consisting of: IGLHDPTQGTEPNGS (SEQ ID NO:56); IGLHDPTQGTEPAGS (SEQ ID NO:57); IGLHDPTQGTLPNGE (SEQ ID NO:58); and IGLHDPTQGTLPAGE (SEQ ID NO:59).
  • the peptide or analog thereof can comprise a peptide or analog selected from: IGLHDPTQGTEPNGS (SEQ ID NO:56); IGLHDPTQGTEPAGS (SEQ ID NO:57); IGLHDPTQGTLPNGE (SEQ ID NO:58); and IGLHDPTQGTLPAGE (SEQ ID NO:59).
  • peptide or analog thereof can consist of: IGLHDPTQGTEPNGS (SEQ ID NO:56); IGLHDPTQGTEPAGS (SEQ ID NO:57); IGLHDPTQGTLPNGE (SEQ ID NO:58); or IGLHDPTQGTLPAGE (SEQ ID NO:59).
  • the peptides or analogs of the invention include analogs of INGAP-PP and HIP that can be peptides having the standard 20 naturally occurring amino acids, as well as other naturally and/or non-naturally occurring amino acids.
  • the peptides as described herein generally use conventional nomenclature. For example, some peptides are designated H-XXX-OH, and it is understood by those skilled in the art that these can designate unmodified amino- (H-) or carboxy- (—OH) termini.
  • the amino acid sequence can also be represented without an indication of a modification on the amino- or carboxy-terminus.
  • peptides described herein can include unmodified and modified amino- and/or carboxy-termini on a peptide comprising a specified amino acid sequence or peptide analog.
  • a peptide or analog comprising a designated amino acid sequence can include additional amino acids on the N- and/or C-terminus as well as modified amino acids of the designated sequence.
  • a peptide or analog comprising a designated peptide or analog similarly can include modified amino acids and/or additional amino acids, unless the N- and/or C-terminus comprises a modification that precludes the addition of an amino acid, for example through a peptide bond.
  • modifications can include, for example, an acetylated N-terminus and/or amidated C-terminus.
  • the peptides or analogs of the invention can comprise a modification. It is understood by those skilled in the art that a number of modifications can be made to a peptide or analog. Exemplary modifications include, but are not limited to, an acetylated N-terminus, an amidated C-terminus, a D amino acid, a modified amino acid, a fatty acid modification, or a combination thereof. Any of a number of well known modifications of a peptide or amino acid can be included in a peptide or analog of the invention. For example, derivatives can include chemical modifications of the polypeptide such as alkylation, acylation, carbamylation, iodination, or any modification which derivatizes the polypeptide. Modifications of a peptide or analog can include modified amino acids, for example, hydroxyproline or carboxyglutamate, and can include amino acids that are not linked by peptide bonds.
  • Chemical synthesis is particularly useful for introducing non-naturally occurring amino acids, modified amino acids and/or a modified N- and/or C-terminus.
  • an advantage of using chemical synthesis to prepare a peptide or analog of the invention is that (D)-amino acids can be substituted for (L)-amino acids, if desired.
  • the incorporation of one or more (D)-amino acids can confer, for example, additional stability of the peptide in vitro or, particularly, in vivo, since endogenous endoproteases generally are ineffective against peptides containing (D)-amino acids.
  • Peptides having D amino acids can also be designated herein using the well known nomenclature of a small letter for the corresponding single letter code for an amino acid.
  • the reactive side group of one or more amino acids in a peptide or analog of the invention can be modified or amino acid derivatives can be incorporated into the peptide.
  • Selective modification of a reactive group of a peptide or analog can impart desirable characteristics upon a peptide or analog. The choice of including such a modification is determined, in part, by the characteristics required of the peptide. For example, a peptide or analog can have a free carboxyl terminus or can be modified so that the C-terminus is amidated (see Tables 2 and 3). Similarly, a peptide or analog can have a free amino terminus or can be modified so that the N-terminus is acetylated (Tables 2 and 3).
  • peptides or analogs of the invention can optionally be amidated on the C-terminus and acetylated on the N-terminus.
  • Other modifications of the N- and/or C-terminus of a peptide or analog can also be included within the meaning of a modification.
  • a peptide or analog of the invention can include, but are not limited to, 2-Aminoadipic acid (Aad); 3-Aminoadipic acid (bAad); beta-Alanine, beta-Aminopropionic acid (bAla); 2-Aminobutyric acid (Abu); 4-Aminobutyric acid, piperidinic acid (4Abu); 6-Aminocaproic acid (Acp); 2-Aminoheptanoic acid (Ahe); 2-Aminoisobutyric acid (Aib); 3-Aminoisobutyric acid (bAib); 2-Aminopimelic acid (Apm); 2,4 Diaminobutyric acid (Dbu); Desmo sine (Des); 2,2′-Diaminopimelic acid (Dpm); 2,3-Diaminopropionic acid (Dpr); N-Ethylglycine (EtGly); N-Ethylasparagine (EtAsn); Hydroxylysine
  • a peptide or analog of the invention includes fatty acid modification.
  • a peptide or analog of the invention can be modified by acylation with aliphatic groups, including C2, C4, C6, C8, C10, C12, C14, C16, C18, C20 or longer chains.
  • the peptide or analog can also be modified by isoprenylation and/or phosphatidylinositol (PI).
  • PI phosphatidylinositol
  • Other amino acid, peptide or protein modifications are well known to those skilled in the art (see, for example, Glazer et al., Chemical modification of proteins: Selected methods and analytical procedures , Elsevier Biomedical Press, Amsterdam (1975)).
  • the invention also includes mimetics of the peptides or analogs disclosed herein, also referred to as peptidomimetics.
  • Mimetics encompass chemicals containing chemical moieties that mimic the function of the peptide. For example, if a peptide contains two charged chemical moieties having functional activity, a mimetic places two charged chemical moieties in a spatial orientation and constrained structure so that the charged chemical function is maintained in three-dimensional space. Thus, a mimetic orients functional groups of a peptide or analog of the invention such that the functional activity of a peptide or analog is retained.
  • Mimetics or peptidomimetics can include chemically modified peptides, peptide-like molecules containing nonnaturally occurring amino acids, peptoids and the like, and have the functional activity of the peptide or analog upon which the peptidomimetic is derived (see, for example, Burger's Medicinal Chemistry and Drug Discovery 5th ed., vols. 1 to 3 (ed. M. E. Wolff; Wiley Interscience 1995)).
  • Methods for identifying a peptidomimetic are well known in the art and include, for example, the screening of databases that contain libraries of potential peptidomimetics (Allen et al., Acta Crystallogr.
  • Mimetics or peptidomimetics can provide desirable properties such as greater stability, for example, when administered to a subject, such as during passage through the digestive tract and, therefore, can be useful for oral administration.
  • mimetics or peptidomimetics are known in the art including, but not limited to, peptide-like molecules which contain a constrained amino acid, a non-peptide component that mimics peptide secondary structure, or an amide bond isostere.
  • a mimetic or peptidomimetic that contains a constrained, non-naturally occurring amino acid can include, without limitation, an ⁇ -methylated amino acid; ⁇ -, ⁇ -dialkylglycine or ⁇ -aminocycloalkane carboxylic acid; an N ⁇ - C ⁇ -cyclized amino acid; an N ⁇ -methylated amino acid; a ⁇ - or ⁇ -amino cycloalkane carboxylic acid; an ⁇ , ⁇ -unsaturated amino acid; a ⁇ , ⁇ -dimethyl or ⁇ -methyl amino acid; a ⁇ -substituted-2,3-methano amino acid; an N-C ⁇ or C ⁇ - C ⁇ cyclized amino acid; a substituted proline or another amino acid mimetic.
  • a mimetic or peptidomimetic which mimics peptide secondary structure can contain, without limitation, a nonpeptidic ⁇ -turn mimic; ⁇ -turn mimic; or mimic of helical structure, each of which is well known in the art.
  • a peptidomimetic also can be a peptide-like molecule which contains an amide bond isostere such as a retro-inverso modification; reduced amide bond; methylenethioether or methylene-sulfoxide bond; methylene ether bond; ethylene bond; thioamide bond; trans-olefin or fluoroolefin bond; 1,5-disubstituted tetrazole ring; ketomethylene or fluoroketomethylene bond or another amide isostere.
  • amide bond such as a retro-inverso modification
  • reduced amide bond reduced amide bond
  • methylenethioether or methylene-sulfoxide bond methylene ether bond
  • ethylene bond thio
  • the invention also provides pseudopeptide derivatives of peptides or analogs of the invention.
  • Pseudopeptides are known in the art as peptides in which a peptide bond (amide bond) in a peptide is modified to an amide bond surrogate (see, for example, Cudic and Stawikowski, Mini - Rev Organic Chem . 4:268-280 (2007); Anderson, in Neuropeptide Protocols , Brent and Carvell, eds. 73:49-60 (1996)).
  • Exemplary amide bond surrogates include, but are not limited to, peptidosulfonamides, phosphonopeptides, depsides and depsipeptides, oliogureas, azapeptides and peptoids (see Cudic and Stawikowski, supra, 2007) as well as as methylene amino, thioether and hydroxyethylene derivatives, and the like (Anderson, supra, 1996).
  • the peptides or analogs of the invention can be produced using methods well known to those skilled in the art, including chemical synthesis of the peptides or analogs using well known methods of peptide synthesis, as described herein. Thus, when the peptides or analogs include one or more non-standard amino acids, it is more likely that they will be produced by a chemical synthetic method. In addition to using chemical synthesis of peptides or analogs, the peptides or analogs can be produced by expression from encoding nucleic acids. This is particularly useful for peptides or analogs that include only naturally occurring amino acids.
  • a nucleic acid encoding the peptide sequence can be prepared using well known methods (see Sambrook et al., Molecular Cloning: A Laboratory Manual , Third Ed., Cold Spring Harbor Laboratory, New York (2001); Ausubel et al., Current Protocols in Molecular Biology , John Wiley and Sons, Baltimore, Md. (1999)). Generally such a nucleic acid will be expressed recombinantly in a suitable host organism such as a bacterium, yeast, mammalian or insect cell, and the like. Production in bacteria can be particularly useful for large scale production of a peptide or analog of the invention. The peptide can be expressed in the organism and purified using well known purification techniques.
  • a nucleic acid molecule encoding the peptide or analog of the invention can be cloned into an appropriate vector, particularly an expression vector, and the encoded peptide or analog can be expressed in a host cell or using an in vitro transcription/translation reaction, thereby providing a means to obtain large amounts of the peptide or analog.
  • the recombinant peptide can be produced as a fusion with a tag, such as a His tag, to facilitate identification and purification.
  • Suitable vectors, host cells, in vitro transcription/translation systems, and tag sequences are well known in the art and commercially available.
  • the peptide or analog can be expressed as a single copy, in a polycistronic expression vector, or optionally can be expressed as a single open reading frame with multiple copies of the peptide sequence.
  • the peptide can be obtained by expressing an open reading frame containing multiple copies of the peptide sequence, resulting in expression of a polypeptide with multiple copies of the peptide.
  • the polypeptide can be post-translationally processed into a peptide or analog of the invention, for example, by engineering appropriate proteolytic cleavage sites between the copies of the peptide and cleaving the polypeptide into the peptide or analog of the invention.
  • a peptide or analog of the invention is a peptide containing only naturally occurring amino acids
  • a method can be employed with expression hosts suitably engineered to express non-naturally occurring amino acids.
  • a peptide or analog expressed recombinantly can optionally be chemically modified to introduce a desired amino acid modification or N- and/or C-terminal modification using well know chemical modification methods (see Glazer et al., supra, 1975).
  • the invention additionally provides nucleic acids encoding peptides or analogs of the invention.
  • nucleic acids include, for example, nucleic acids encoding any of the amino acid sequences of SEQ ID NOS:6-73.
  • the analogs when the analogs include only one or more substitutions with standard amino acids, the analogs can be expressed from an expression vector using well known methods, as disclosed herein.
  • the peptides or analogs of the invention can comprise a sequence or peptide or analog as disclosed herein.
  • the peptide will generally have a length of 20 amino acids or less.
  • the peptide or analog can have a length of 19 amino acids or less, 18 amino acids or less, 17 amino acids or less.
  • a peptide or analog of the invention, as disclosed herein can have a length of 10 amino acids, 11 amino acids, 12 amino acids, 13 amino acids 14 amino acids (see Peptide 73, Peptide 74), 15 amino acids, 16 amino acids, 17 amino acids, 18 amino acids, 19 amino acids, or 20 amino acids.
  • shorter peptides includes a fragment of a disclosed peptide or analog, for example, by deletion of one or more amino acids on the N- and/or C-terminus of a disclosed peptide or analog, that retains functional activity, including but not limited to one or more of the biological activities of peptides and analogs of the invention, as disclosed herein. Nevertheless, it is understood that a peptide can also comprise longer amino acid lengths, so long as the functional activity of the peptide or analog is retained.
  • a peptide or analog can have a length of less than 150 residues, less than 130 residues, less than 120 residues, less than 110 residues, less than 100 residues, less than 90 residues, less than 80 residues, less than 70 residues, less than 60 residues, less than 50 residues, less than 45 residues, less than 40 residues, less than 35 residues, less than 30 residues, less than 25 residues, less than 24 residues, less than 23 residues, less than 22 residues, less than 21 residues, less than 20 residues, less than 19 residues, less than 18 residues, or less than 17 residues. It is understood by those skilled in the art that, where a peptide or analog of the invention comprises a sequence found within a known longer sequence such as a wild type full length protein, the peptide or analog of the invention specifically excludes such a full length sequence.
  • the invention also provides peptides and analogs of the invention in a pharmaceutically acceptable salt form that is well known to those skilled in the art.
  • a particularly useful salt form is acetate or hydrochloride salt form. Nevertheless, it is understood by those skilled in the art that any of a number of suitable salt forms are available.
  • the peptide or analog of the invention contains an acidic or basic moiety, it can be provided as a pharmaceutically acceptable salt (see, for example, Berge et al., J. Pharm. Sci. 1977, 66, 1-19; and Handbook of Pharmaceutical Salts, Properties, and Use ; Stahl and Wermuth, Ed.; Wiley-VCH and VHCA: Zurich, Switzerland, 2002).
  • Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+)-camphoric acid, camphorsulfonic acid, (+)-(1S)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid,
  • Suitable bases for use in the preparation of pharmaceutically acceptable salts including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2-(diethylamino)-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, N-methyl-glucamine, hydrabamine, 1H-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, 1-(2-hydroxyethyl
  • the invention also provides peptides and analogs of the invention in a composition.
  • the composition can optionally be formulated with a pharmaceutically acceptable carrier to produce a pharmaceutical composition, which can be administered to the individual, which can be a human or other mammal.
  • a pharmaceutically acceptable carrier can be, for example, water, sodium phosphate buffer, phosphate buffered saline, normal saline or Ringer's solution or other physiologically buffered saline, or other solvent or vehicle such as a glycol, glycerol, an oil such as olive oil or an injectable organic ester.
  • a pharmaceutically acceptable carrier can contain physiologically acceptable compounds that act, for example, to stabilize or increase the absorption of the peptide or analog of the invention.
  • physiologically acceptable compounds include, for example, carbohydrates such as glucose, sucrose or dextrans; antioxidants such as ascorbic acid or glutathione; chelating agents such as ethylenediamine tetraacetic acid (EDTA), which disrupts microbial membranes; divalent metal ions such as calcium or magnesium; low molecular weight proteins; or other stabilizers or excipients.
  • EDTA ethylenediamine tetraacetic acid
  • Suitable carriers and their formulations are well known in the art (see, for example, Remington: The Science and Practice of Pharmacy, 19th ed., ed. A. R. Gennaro, Mack Publishing Company, Easton, Pa. (1995); and Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton Pa. (1990)).
  • an appropriate amount of a pharmaceutically-acceptable salt is used in the formulation to render the formulation isotonic.
  • the pH of the solution is generally from about 5 to about 8, for example, from about 7 to about 7.5.
  • Pharmaceutical carriers are known to those skilled in the art. These most typically would be standard carriers for administration of drugs to humans, including solutions such as sterile water, saline, and buffered solutions at physiological pH, as described above.
  • Pharmaceutical compositions can include carriers, thickeners, diluents, buffers, preservatives, surface active agents and the like in addition to the molecule of choice such as the peptides or analogs of the invention.
  • Pharmaceutical compositions can also include one or more active ingredients such as antimicrobial agents, anti-inflammatory agents, anesthetics, and the like.
  • Further carriers include sustained or controlled release preparations such as semipermeable matrices of solid hydrophobic polymers covalently or non-covalently bound to the peptide or analog, which matrices are in the form of shaped articles, for example, films, liposomes, non-liposome lipid complex or microparticles, and the like, or other biocompatible polymers well known to those skilled in the art (see, for example, U.S. Pat. Nos. 6,824,822 and 8,329,648).
  • Liposomes which consist of phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer (Gregoriadis, Liposome Technology , Vol.
  • the pharmaceutical composition can be administered in a number of ways depending on whether local or systemic treatment is desired, and on the area to be treated. It is understood that a variety of routes of administration are useful for the peptides, analogs and methods of the invention. Such routes encompass systemic and local administration and include, without limitation, intravenous injection, intraperitoneal injection, intramuscular injection, subcutaneous injection, transdermal delivery, transdermal diffusion or electrophoresis, inhalable administration, oral administration, local injection, intracavity, and extended release delivery devices including locally implanted extended release devices such as bioerodible or reservoir-based implants. Administration can be topically (including ophthalmically, vaginally, rectally, intranasally), orally, by inhalation, or parenterally, for example by intravenous drip, subcutaneous, intraperitoneal or intramuscular injection.
  • Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives can also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like.
  • Insulin is a well known peptide therapeutic, so methods used for delivery of insulin are particularly amenable as a delivery method for peptides or analogs of the invention, including but not limited to syringes, pens, infusion pumps, inhalers, mouth sprays, pills, and the like.
  • the peptides and analogs of the invention are particularly useful for treating certain diseases and disorders.
  • the peptides or analogs of the invention can be used for treating impaired pancreatic function, treating a metabolic disease, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation.
  • the invention additionally provides compositions of the invention for treating impaired pancreatic function, treating a metabolic disease, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation.
  • the use of the peptides and analogs of the invention in such therapeutic applications are described below in more detail.
  • the peptides or analogs of the invention can be administered in combination.
  • a combination of two or more peptides or analogs of the invention can be administered for a method of treatment as disclosed herein.
  • Such a combination can be administered concurrently, either in separate formulations or combined into the same formulation, depending on the peptides being administered and the compatibility of the formulations for the peptides or analogs of the invention.
  • the two or more peptides or analogs of the invention can be administered sequentially, including on the same day or staggered on separate days.
  • the peptide and analogs of the invention can optionally be administered with drugs or therapeutic agents for treating a condition.
  • drugs or therapeutic agents for treating a condition.
  • other anti-diabetic drugs can be administered with the peptides or analogs of the invention.
  • co-administration can occur concurrently, either in separate formulations or combined into the same formulation, depending on the drugs being administered and the compatibility of the formulations for the peptides or analogs of the invention.
  • the co-administration can occur sequentially, including on the same day or staggered on separate days.
  • appropriate administration regimens suitable for effective deliver of a peptide or analog of the invention with another drug or therapeutic agent are examples of administration regimens suitable for effective deliver of a peptide or analog of the invention with another drug or therapeutic agent.
  • suitable anti-diabetic drugs include, but are not limited to, insulin, pramlintide, GLP-1 receptor agonists, oral anti-diabetic agents, and the like.
  • exemplary anti-diabetic drugs include, but are not limited to, insulin, meglitinides, for example, repaglinide (PrandinTM) and nateglinide (StarlixTM); sulfonylureas, for example, glipizide (GlucotrolTM), glimepiride (AmarylTM), and glyburide (DiaBetaTM, GlynaseTM); dipeptidy peptidase-4 (DPP-4) inhibitors, for example, saxagliptin (OnglyzaTM), sitagliptin (JanuviaTM), and linagliptin (TradjentaTM); biguanides, for example, metformin (FortametTM, GlucophageTM);
  • Type 1 diabetes and latent autoimmune diabetes in adults are both autoimmune diseases. Therefore, in the case of a subject having type 1 diabetes or LADA, another therapeutic agent that can be administered with a peptide or analog of the invention can be, for example, an immune modulatory agent.
  • the immunomodulatory agent can be used to block or reduce the destruction of neogenic islet or beta cells associated with autoimmunity.
  • Exemplary immunodulatory agents include, but are not limited to, sirolimus (rapamycin, RapamuneTM), tacrolimus (FK 506, PrografTM), lisofylline, antithymocyte globulin, basiliximab (SimulectTM), DiaPep277TM, and the like.
  • the peptides and analogs of the invention exhibit unexpected properties over that of the parent INGAP-PP and HIP peptides, including the peptides and analogs of Tables 2 and 3 that are not the parent INGAP-PP or HIP peptides.
  • peptides and analogs of the invention exhibit improved stability in culture medium and plasma over that of the parent peptide (see Example III).
  • the peptide analogs of the invention also were effective at significantly improving blood glucose, fasting insulin and oral glucose tolerance (see Example IV).
  • the peptide analogs of the invention also exhibit a significantly increased islet neogenic effect than the parent peptide (see Example V).
  • the peptide analogs of the invention exhibit a significantly increased ability to stimulate insulin secretion in primary pancreatic islet cells (see Example VI). Additionally, peptide analogs of the invention exhibited superior pharmacokinetic properties (see Example VII). The numerous unexpected and superior properties of the peptides and analogs of the invention indicate that the peptides and analogs of the invention, including the peptides and analogs of Tables 2 and 3 that are not the parent INGAP-PP or HIP peptides, can be utilized for therapeutic applications.
  • the invention provides a method for ameliorating a sign or symptom associated with impaired pancreatic function comprising administering a peptide or analog of the invention.
  • a disease or condition associated with impaired pancreatic function includes, but is not limited to, type 1 diabetes, type 2 diabetes, latent autoimmune diabetes in adults (LADA), impaired fasting glucose, impaired glucose tolerance, insulin deficiency, fasting hyperinsulinemia, insulin resistance, or impaired fasting insulin levels, or a combination thereof.
  • LADA latent autoimmune diabetes in adults
  • impaired fasting glucose impaired glucose tolerance
  • insulin deficiency insulin deficiency
  • fasting hyperinsulinemia insulin resistance
  • impaired fasting insulin levels or a combination thereof.
  • the pancreas produces insulin for regulation of blood glucose.
  • Diabetes mellitus is a serious metabolic disease that is defined by the presence of chronically elevated levels of blood glucose (hyperglycemia). This state of hyperglycemia is the result of a relative or absolute lack of activity of the peptide hormone, insulin.
  • Insulin is produced and secreted by the ⁇ -cells of the pancreas. Insulin promotes glucose utilization, protein synthesis, and the formation and storage of carbohydrate energy as glycogen.
  • Glucose is stored in the body as glycogen, a form of polymerized glucose, which may be converted back into glucose to meet metabolism requirements. Under normal conditions, insulin is secreted at both a basal rate and at enhanced rates following glucose stimulation, all to maintain metabolic homeostasis by the conversion of glucose into glycogen.
  • diabetes mellitus encompasses several different hyperglycemic states. These states include type 1 (insulin-dependent diabetes mellitus or IDDM) and type 2 (non-insulin dependent diabetes mellitus or NIDDM) diabetes.
  • IDDM insulin-dependent diabetes mellitus
  • NIDDM non-insulin dependent diabetes mellitus
  • the hyperglycemia present in individuals with type 1 diabetes is associated with deficient, reduced, or nonexistent levels of insulin which are insufficient to maintain blood glucose levels within the physiological range.
  • Treatment of type 1 diabetes involves administration of replacement doses of insulin, generally by a parenteral route.
  • the hyperglycemia present in individuals with type 2 diabetes is initially associated with normal or elevated levels of insulin; however, these individuals are unable to maintain metabolic homeostasis due to a state of insulin resistance in peripheral tissues and liver and, as the disease advances, due to a progressive deterioration of the pancreatic ⁇ cells which are responsible for the secretion of insulin.
  • initial therapy of type 2 diabetes may be based on diet and lifestyle changes augmented by therapy with oral hypoglycemic agents such as sulfonylureas. Insulin therapy is often required, however, especially in the latter states of the disease, in order to produce some control of hyperglycemia and minimize complications of the disease.
  • the invention additionally provides a method for ameliorating a sign or symptom associated with a metabolic disease in a subject comprising administering a peptide or analog of the invention to the subject.
  • a metabolic disease includes, but is not limited to, diabetes, pre-diabetes or metabolic syndrome.
  • Prediabetes is a condition where blood sugar level is higher than normal but not yet high enough to be classified as type 2 diabetes.
  • Metabolic syndrome is a name for a group of risk factors that occur together and increase the risk for coronary artery disease, stroke, and type 2 diabetes.
  • the two most important risk factors for metabolic syndrome are extra weight around the middle and upper parts of the body (central obesity)(so-called “apple-shaped”) and insulin resistance, where the body uses insulin less effectively than normal. Insulin is needed to help control the amount of sugar in the body. As a result, blood sugar and fat levels rise.
  • Metabolic syndrome is considered to be present if a subject has three or more of the following signs: blood pressure equal to or higher than 130/85 mmHg; fasting blood sugar (glucose) equal to or higher than 100 mg/dL; large waist circumference (length around the waist)(men, 40 inches or more; women, 35 inches or more); low HDL cholesterol (men, under 40 mg/dL; women, under 50 mg/dL); triglycerides equal to or higher than 150 mg/dL.
  • blood pressure equal to or higher than 130/85 mmHg
  • fasting blood sugar (glucose) equal to or higher than 100 mg/dL
  • large waist circumference length around the waist
  • low HDL cholesterol men, under 40 mg/dL; women, under 50 mg/dL
  • triglycerides equal to or higher than 150 mg/dL.
  • both type 1 and type 2 diabetes are a well characterized diseases with a number of known parameters for diagnosing and/or monitoring the progression of the disease and/or to monitor the effectiveness of a therapy.
  • Such parameters include, but are not limited to, plasma glucose levels, fasting glucose levels, oral glucose tolerance test (OGTT), insulin levels, fasting insulin levels, glycosylated hemoglobin levels, and the like.
  • the peptides or analogs of the invention can therefore be used to ameliorate any one or more of the signs or symptoms associated with impaired pancreatic function and/or metabolic disease.
  • signs or symptoms include, but are not limited to, impaired glucose tolerance, increased blood glucose (in particular above 200 mg/dl), increased fasting blood glucose (in particular above 140 mg/dl), increased postprandial (after eating) blood glucose, insulin deficiency, fasting hyperinsulinemia, insulin resistance, impaired fasting insulin levels, increased glycosylated hemoglobin (HbA1c), and the like.
  • Such signs or symptoms are well known to those skilled in the art and can be routinely determined by those skilled in the art, including tests available through medical testing laboratories.
  • the invention provides a method of reducing a sign or symptom associated with a condition such as diabetes, for example, a method of reducing impaired glucose tolerance, blood glucose, in particular daily average blood glucose concentration, fasting blood glucose, postprandial (after eating) blood glucose, insulin deficiency, fasting hyperinsulinemia, insulin resistance, impaired fasting insulin levels, glycosylated hemoglobin (HbA1c), arginine-stimulated C-peptide, advanced glycation end products (AGE), or a combination thereof, by administering a peptide or analog of the invention.
  • a condition such as diabetes
  • a method of reducing impaired glucose tolerance, blood glucose in particular daily average blood glucose concentration, fasting blood glucose, postprandial (after eating) blood glucose, insulin deficiency, fasting hyperinsulinemia, insulin resistance, impaired fasting insulin levels, glycosylated hemoglobin (HbA1c), arginine-stimulated C-peptide, advanced glycation end products (AGE), or
  • the invention provides a method of reducing in a diabetic subject impaired glucose tolerance, blood glucose, fasting blood glucose, postprandial blood glucose, insulin deficiency, fasting hyperinsulinemia, insulin resistance, impaired fasting insulin levels, glycosylated hemoglobin (HbA1c), arginine-stimulated C-peptide, advanced glycation end products (AGE), or a combination thereof, by administering a peptide or analog of the invention to the subject.
  • HbA1c glycosylated hemoglobin
  • AGE advanced glycation end products
  • the peptides and analogs of the invention were particularly effective at stimulating pancreatic islet cell growth and induction of ⁇ -cell clusters (see Example V).
  • Exemplary peptides and analogs of the invention exhibited improved islet neogenic effect over parent peptide (Example V).
  • the invention additionally provides a method for stimulating pancreatic islet cell growth by contacting a pancreatic islet cell in vitro with a peptide or analog of the invention, whereby proliferation of the pancreatic islet cell is stimulated.
  • the invention provides a method of producing a population of pancreatic islet cells, comprising contacting one or more pancreatic islet cells in vitro with a peptide or analog of the invention, whereby proliferation of the one or more pancreatic islet cells is stimulated and a population of pancreatic islet cells is produced.
  • the methods of the invention can be used for ex vivo islet induction, expansion and proliferation for transplantation and for increasing the survival of transplanted islets in vivo.
  • the invention provides methods of ex vivo islet expansion and proliferation for transplantation using the peptides or analogs of the invention by contacting islet cells in vitro, increasing the islet cell numbers and optionally using the cells for transplantation.
  • the invention also provides a method of increasing the survival of transplanted islets in vivo by administering to a subject a peptide or analog of the invention, wherein the subject is the recipient of transplanted islet cells.
  • the peptides or analogs of the invention can thus be used to generate cells for transplantation using in vitro and ex vivo methods as well as to increase survival of transplanted islet cells.
  • Such transplanted cells can be obtained from the in vitro methods using the peptides or analogs of the invention or from traditional transplant sources of islet cells such as cadavers.
  • the one or more pancreatic islet cells can be obtained from a subject.
  • the population of pancreatic islet cells produced by stimulating proliferation of the pancreatic islet cells can be used, for example, for transplantation into a subject and restoration of pancreatic islet cell function.
  • a method of the invention can further comprise the step of transplanting the population of pancreatic islet cells into a subject.
  • the one or more pancreatic cells are obtained from the subject into which the population of pancreatic islet cells is to be transplanted.
  • the pancreatic islet cells to be transplanted are obtained from a suitable donor having a compatible blood type.
  • pancreatic islet cells can be obtained from the subject or, alternatively, from a suitable donor, including islet cells harvested from a cadaver.
  • the transplant recipient is administered immunosuppressive drugs to decrease rejection of the islet cells (see, for example, immunosuppressive drugs described herein).
  • immunosuppressive drugs are well known in the field of organ or cell transplantation.
  • pancreatic islet cells in methods of the invention in which pancreatic islet cells are stimulated to proliferate in vitro to produce a population of pancreatic islet cells, such a population can be transplanted into a subject using well known methods of pancreatic islet cell transplantation.
  • peptides or analogs of the invention can be used to induce differentiation of pancreatic ductal cells into islet cells, in particular beta-cells (see Yatoh et al., Diabetes 56:1802-1809 (2007)).
  • the invention further provides a method of differentiating pancreatic ductal cells into islet cells by contacting a pancreatic ductal cell with a peptide or analog of the invention. When the method is performed where the pancreatic ductal cell is contacted in vitro, a population of differentiated pancreatic ductal cells can be generated and used for transplantation, as described herein.
  • the invention further provides a method for increasing the number of pancreatic islet cells in a subject comprising administering a peptide or analog of the invention to the subject.
  • a method of therapeutic treatment using peptides or analogs of the invention can be used to increase pancreatic islet cells in an individual, without the need to harvest pancreatic cells from the individual or identify a suitable donor and without the need to put the subject through complex transplantation procedures and the frequently required use of immunosuppressive agents if using donor cells not obtained from the patient.
  • INGAP peptide has been shown to improve nerve function and enhance nerve regeneration in a diabetic mouse model (Tam et al., FASEB J. 18:1767-1769 (2004)). INGAP peptide was also shown to enhance neurite outgrowth in dorsal root ganglia neurons (Tam et al., Biochem. Biophys. Res. Communic. 291:649-654 (2002; Tam et al., NeuroReport 17:189-193 (2006)). As described herein, the peptides and analogs of the invention are significantly more active than the INGAP parent peptide and are expected to have a similar but more potent activity than INGAP.
  • the invention provides a method for promoting neuroprotection or nerve regeneration by contacting a nerve cell with a peptide or analog the invention, thereby stimulating neuroprotection and/or nerve regeneration.
  • the contacting with a nerve cell can occur in vivo or in vitro.
  • the peptide or analog of the invention is administered to a subject as with other therapeutic methods disclosed herein.
  • the neuroprotected cell can be used in an ex vivo application and the cell administered to the subject.
  • Such methods of introducing nerve cells by way of transplantation are well known to those skilled in the art (see, for example, Dunnett et al., Brit. Med. Bulletin 53:757-776 (1997)). Such transplantations have been performed to treat neurological conditions such as Parkinson's disease and Huntington's disease.
  • the HIP peptide has been described as accelerating liver regeneration (Lieu et al., Hepatol. 42:618-626 (2005).
  • the peptides and analogs of the invention are significantly more active than the HIP parent peptide and are expected to have a similar but more potent activity than HIP.
  • the invention also provides a method for promoting liver regeneration by contacting a liver cell with a peptide or analog of the invention, thereby promoting liver regeneration.
  • the contacting with a liver cell can occur in vivo or in vitro.
  • the peptide or analog of the invention is administered to a subject as with other therapeutic methods disclosed herein.
  • the liver cells can be induced to proliferate, for example, to produce a population of liver cells.
  • the population of liver cells can be used in an ex vivo application and the cells administered to the subject.
  • Methods for transplanting or grafting liver cells onto the liver of a subject are well known to those skilled in the art.
  • the transplanted cells can be used to reconstitute injured, or metabolically defective, liver tissue.
  • Liver cells can be infused into the portal vein or spleen from where cells migrate to the liver and take up permanence residence and perform the normal liver metabolic functions (see, for example, Khan et al., Cell Transplant. 19:409-418 (2010)).
  • the HIP protein also referred to as Pancreatitis-associated protein (PAP)
  • PAP Pancreatitis-associated protein
  • the invention also provides the use of a peptide or analog of the invention for preparation of a medicament for treating impaired pancreatic function, treating a metabolic disease, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation in a subject.
  • the invention additionally provides use of a peptide or analog of the invention for preparation of a medicament for treating impaired pancreatic function, treating a metabolic disease, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation in a subject.
  • a peptide or analog of the invention for preparation of a medicament for treating impaired pancreatic function, treating a metabolic disease, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation in a subject.
  • Such uses can be, for example, to carry out the methods of the invention disclosed herein.
  • the peptides and analogs of the invention can be used in a variety of methods. Such methods include, but not limited to, treating impaired pancreatic function, treating a metabolic disease, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation. In many applications of the invention for a therapeutic application, the peptides or analogs of the invention are administered. However, it is understood that an alternative mode is to use gene therapy to express a peptide of the invention by administering a suitable gene therapy vector containing a nucleic acid encoding the peptide to a subject. Such gene therapy methods are described below in more detail and are well known to those skilled in the art (see, for example, Anderson, Nature 392 (Supp.):25-30 (1998)).
  • a gene delivery vehicle refers to a molecule that can carry inserted polynucleotides into a host cell.
  • gene delivery vehicles are liposomes, micells biocompatible polymers, including natural polymers and synthetic polymers; lipoproteins; polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; metal particles; and bacteria, or viruses, such as baculovirus, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression.
  • a peptide or analog of the invention can be delivered to a cell or tissue using a gene delivery vehicle.
  • Gene delivery, gene transfer, transducing, and the like as used herein, are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a transgene) into a host cell, irrespective of the method used for the introduction.
  • exogenous polynucleotide sometimes referred to as a transgene
  • Such methods include a variety of well-known techniques such as vector-mediated gene transfer (by, e.g., viral infection/transfection, or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of “naked” polynucleotides (such as electroporation, “gene gun” delivery and various other techniques used for the introduction of polynucleotides).
  • the introduced polynucleotide can be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome.
  • a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome.
  • a number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art.
  • a viral vector refers to a recombinantly produced virus or viral particle that comprises a polynucleotide to be delivered into a host cell, either in vivo, ex vivo or in vitro.
  • viral vectors include retroviral vectors, adenovirus vectors, adeno-associated virus vectors, alphavirus vectors and the like.
  • Alphavirus vectors such as Semliki Forest virus-based vectors and Sindbis virus-based vectors, have also been developed for use in gene therapy and immunotherapy (see Schlesinger and Dubensky Curr. Opin. Biotechnol. 5:434-439 (1999) and Ying, et al. Nat. Med. 5(7):823-827 (1999)).
  • a vector construct refers to the polynucleotide comprising the retroviral genome or part thereof, and a therapeutic gene.
  • retroviral mediated gene transfer or retroviral transduction carries the same meaning and refers to the process by which a gene or nucleic acid sequences are stably transferred into the host cell by virtue of the virus entering the cell and integrating its genome into the host cell genome.
  • the virus can enter the host cell via its normal mechanism of infection or be modified such that it binds to a different host cell surface receptor or ligand to enter the cell.
  • retroviral vector refers to a viral particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism. Retroviruses carry their genetic information in the form of RNA; however, once the virus infects a cell, the RNA is reverse-transcribed into the DNA form which integrates into the genomic DNA of the infected cell. The integrated DNA form is called a provirus.
  • a vector construct refers to the polynucleotide comprising the viral genome or part thereof, and a transgene.
  • Ads adenoviruses
  • Ads are a relatively well characterized, homogenous group of viruses, including over 50 serotypes (see, for example, WO 95/27071). Ads do not require integration into the host cell genome. Recombinant Ad derived vectors, particularly those that reduce the potential for recombination and generation of wild-type virus, have also been constructed (see, for example, WO 95/00655 and WO 95/11984).
  • Wild-type AAV has high infectivity and specificity integrating into the host cell's genome (see, for example, Hermonat and Muzyczka, Proc. Natl. Acad. Sci. USA 81:6466-6470 (1984) and Lebkowski et al., Mol. Cell. Biol. 8:3988-3996 (1988)).
  • Vectors that contain both a promoter and a cloning site into which a polynucleotide can be operatively linked are well known in the art. Such vectors are capable of transcribing RNA in vitro or in vivo, and are commercially available from sources such as Stratagene (La Jolla, Calif.) and Promega Biotech (Madison, Wis.). In order to optimize expression and/or in vitro transcription, it may be necessary to remove, add or alter 5 ‘ and/or 3’ untranslated portions of the clones to eliminate extra, potential inappropriate alternative translation initiation codons or other sequences that may interfere with or reduce expression, either at the level of transcription or translation. Alternatively, consensus ribosome binding sites can be inserted immediately 5′ of the start codon to enhance expression.
  • Gene delivery vehicles also include DNA/liposome complexes, micelles and targeted viral protein-DNA complexes. Liposomes that also comprise a targeting antibody or fragment thereof can be used in the methods of this invention.
  • the nucleic acid or proteins of this invention can be conjugated to antibodies or binding fragments thereof which bind cell surface antigens, for example, a cell surface marker found on pancreatic islet cells.
  • the invention provides a method of introducing a peptide or analog of the invention into a subject by contacting a cell with a nucleic acid encoding a peptide or analog of the invention.
  • the contacting of a cell with the nucleic acid can occur in vitro, for ex vivo applications, or in vivo. Such methods are often referred to as gene therapy methods.
  • the cells expressing the polynucleotide can be administered to the subject.
  • Such methods permit the expression of a therapeutic protein or peptide, such as the peptides or analogs of the invention, for therapeutic applications.
  • Such therapeutic applications can be used for treating various diseases and conditions, including but not limited to treating impaired pancreatic function, treating a metabolic disease, promoting neuroprotection or nerve regeneration, promoting liver regeneration or inhibiting inflammation, as disclosed herein.
  • This example describes the production of peptides and peptide analogs.
  • Chain elongation reaction was performed followed by Fmoc deprotection in 20% piperidine in DMF.
  • the Fmoc protecting groups were removed from the N terminus of the peptides by 25% piperidine in DMF followed by washing with DMF for four times.
  • TFA trifluoroacetic acid
  • a solution of 20% acetic anhydride dissolved in DMF was added at a ratio of 7 mL/g resin, reacted for 30 mins, followed by 4 times washes with DMF and DCM. Following washing for four times with DMF and DCM, the resin was dried under vacuum.
  • the prepared peptides were cleaved from the resin using standard TFA cleavage procedures in TFA with 5% H 2 O followed by multiple ether extractions. All synthetic peptides were purified to >95% by reverse-phase high-pressure liquid chromatography performed with a liquid chromatograph. Peptides were analyzed by mass spectrometry to confirm the identity and purity.
  • the above prepared peptides were dissolved in double distilled water to make a stock solution, and in the in vivo efficacy study they were reconstituted in sterile normal saline to reach the desired concentration.
  • the final peptide solution was filtered through a 0.22 ⁇ m membrane to make it sterile.
  • the peptides and analogs can also be produced using other well known methods, including manufacturing the peptides using a method of peptide synthesis or expressing nucleic acids that code for the desired peptides or peptide analogs.
  • the analogs include one or more non-standard amino acids, it is more likely that they will be produced by a chemical synthetic method.
  • the peptides include only one or more substitutions with standard amino acids, the peptides can be expressed from an expression vector using well known expression methods.
  • This example describes the effect of peptides and analogs on pancreatic cell growth.
  • a bromodeoxyuridine (BrdU) ELISA assay was performed. Briefly, ARIP cells (ATCC (American Type Culture Collection), Manassas Va. USA), a rat pancreatic ductal cell line, were cultured in F-12K medium (Gibco-BRL, Gaithersburg, Md., USA) containing 10% fetal bovine serum (FBS; HyClone, Thermo Fisher Scientific Inc.; Waltham Mass. USA), 100 ⁇ g/ml streptomycin and 100 ⁇ g/ml penicillin in a cell incubator.
  • FBS HyClone, Thermo Fisher Scientific Inc.
  • penicillin 100 ⁇ g/ml penicillin
  • ARIP cells were seeded into 96-well culture plates at 8000 or 0 (as blank control) cells/well in a volume of 50 ⁇ l cell culture medium and incubated overnight for the following experiments.
  • 50 ⁇ l serum-free cell culture medium containing test peptides at a series concentrations were added to seeded cells.
  • Medium without compound was added to negative control and background control wells. The medium was replaced at 24 h and 48 h respectively with fresh medium.
  • the medium was supplemented with 10 ⁇ l bromodeoxyuridine (BrdU) labeling solution (except the background control wells) from a BrdU cell proliferation ELISA kit (Roche Applied Science; Indianapolis Ind. USA), and incubated for an additional 3 hours.
  • labeling medium was removed, and 200 Owen of FixDenat solution was added.
  • FixDenat solution was removed thoroughly and 100 ⁇ l/well of anti-BrdU antibody working solution was added and incubated at room temperature (RT) for 90 minutes.
  • RT room temperature
  • Antibody conjugate was removed and wells were rinsed three times with 250 ⁇ l/well Washing solution (1 ⁇ PBS).
  • ARIP cells ATCC, cat# CRL-1674 were cultured in F-12K medium (Gibco-BRL) containing 10% fetal bovine serum (FBS; HyClone), 100 ⁇ g/ml streptomycin and 100 ⁇ g/ml penicillin in a cell incubator.
  • FBS fetal bovine serum
  • ARIP cells were seeded into 96-well culture plates at 8000 and 0 (as blank control) cells/well in the volume of 50 ⁇ l cell culture medium and incubated overnight for the following experiments.
  • FIG. 1 shows the comparison of ARIP cell proliferation in the presence of 100 nM of INGAP Scrambled PP 1 (Peptide 3), INGAP-PP (Peptide 1), and Peptide 7 (peptides shown in Table 2).
  • FIG. 1 shows that there was an increase in cell number at a peptide concentration of 100 nM.
  • Peptide 7 showed a significantly higher percentage increase in cell number compared to the INGAP scrambled peptide, a negative control, and INGAP-PP peptide.
  • This example describes stability studies of peptides in various conditions.
  • a certain amount of selected peptides was accurately weighed and dissolved in distilled water to 5 mg/mL as a stock solution.
  • the stock solution was diluted to 0.25 mg/mL with F-12K medium (Gibco-BRL, Gaithersburg, Md., USA) as working solution.
  • F-12K medium Gibco-BRL, Gaithersburg, Md., USA
  • a volume of 100 ⁇ L of each working solution was transferred into individual sample vials.
  • the sample vials were incubated in a 37° C. incubator for 0, 24, 48 and 72 hours before being analyzed and quantitated by HPLC.
  • FIG. 2 shows the stability of compounds in culture medium.
  • FIG. 2 shows a stability comparison in culture medium of INGAP-PP (Peptide 1) and selected peptide analogs, Peptide 7 and Peptide 8 (see Table 2).
  • peptide analogs Peptide 7 and Peptide 8 were significantly more stable than INGAP-PP peptide in culture medium.
  • peptides and eucatropine powder were accurately weighed.
  • Test compounds were dissolved in 50% methanol-water solution and diluted to 20 mg/mL, and eucatropine was dissolved in dimethylsulfoxide (DMSO) and diluted to 10 mM, as a stock solution.
  • Eucatropine stock solution was diluted to 0.2 mM with DMSO as a working solution.
  • a stop reagent was prepared containing 200 ng/mL midazolam and tolbutamide in acetonitrile. A volume of 300 ⁇ L of stop solution was added to each well of a 96-well deep-well plate placed on ice beforehand.
  • FIG. 3 shows stability of compounds in mouse plasma.
  • FIG. 3 shows a stability comparison in mouse plasma of INGAP-PP (Peptide 1) and selected peptide analogs, Peptide 12, Peptide 16 and Peptide 29 (see Table 2).
  • INGAP-PP Peptide 1
  • Peptide 16 and Peptide 29 selected peptide analogs, Peptide 12, Peptide 16 and Peptide 29 (see Table 2).
  • peptide analogs Peptide 12, Peptide 16 and Peptide 29 exhibited good stability in mouse plasma and were more stable than INGAP-PP (Peptide 1).
  • FIG. 5 shows stability of compounds in mouse plasma.
  • FIG. 5 shows a stability comparison in mouse plasma of HIP (Peptide 2) and selected peptide analogs, Peptide 52 and Peptide 54 (see Table 3).
  • peptide analogs Peptide 52 and Peptide 54 exhibited good stability in mouse plasma and were significantly more stable than HIP (Peptide 2).
  • FIG. 4 shows the stability of compounds in human plasma.
  • FIG. 4 shows a stability comparison in human plasma of INGAP-PP (Peptide 1) and selected peptide analogs, Peptide 12 and Peptide 16 (see Table 2).
  • peptide analogs Peptide 12 and Peptide 16 exhibited good stability in human plasma and were significantly more stable than INGAP-PP (Peptide 1).
  • This example describes an in vivo efficacy study using a streptozotocin (STZ) induced diabetic mice model.
  • mice After acclimatization in the animal facility for one week, 6-8 weeks old C57BL/6J mice were administered low dose STZ at 40 mg/kg in citrate buffer for 5 consecutive days to establish a T1D animal model. Mice with blood glucose greater than 16.7 mmol/L at 5 days post last STZ injection were included in the study. These mice were then treated with INGAP-PP (Peptide 1) or Peptide 7 at the doses of 5 mg/kg (2.5 mg/kg, bid (twice a day)) or 25 mg/kg (12.5 mg/kg, bid) for 20 days before sacrifices.
  • INGAP-PP Peptide 1
  • Peptide 7 25 mg/kg (12.5 mg/kg, bid
  • mice Two additional groups of diabetic mice were administered either saline or a peptide (Peptide 3) composed of a scrambled sequence of amino acids from Peptide 1 as control groups.
  • Blood glucose and insulin levels were measured, and 20 days post the last dosing of test agents, an oral glucose tolerance test (OGTT) was performed in 6 hour fasted animals to determine the effect of Peptide 1 and Peptide 7.
  • Plasma samples obtained from the tail cut for glucose determination were detected with an ACCU-CHEKTM glucometer (Roche, ACCU-CHEK® Active), and insulin levels were determined with Rat/Mouse Insulin Elisa kit (Millipore, Billerica, Mass. USA).
  • mice received an oral glucose challenge at 2 g/kg and glucose values were determined by glucometer at 0, 15, 30, 60, 90 and 120 min.
  • FIG. 6 shows the efficacy comparison of INGAP-PP (Peptide 1), INGAP Scrambled PP 1 (Peptide 3) and Peptide 7 in STZ induced diabetic mice model.
  • FIG. 6A shows the blood glucose (BG, mM) on day 21 of treatment.
  • FIG. 6B shows the fasting insulin levels (ng/ml) on day 21 of treatment.
  • FIG. 6C shows the area under curve (AUC) of glucose (T 0-120 min ) measured in an oral glucose tolerance test (OGTT) on day 21 of treatment.
  • This example describes the effects of peptides on the induction of small ⁇ -cell clusters in normal C57BL/6J mice.
  • mice were randomly divided into 4 groups.
  • Body weight and 6 hour fasting blood glucose were measured before treatment and after the last dosing of treatment. Plasma and pancreatic insulin were also measured at the end of the study. On day 11, the pancreas was removed from each animal, cleared of fat and lymph nodes, weighed, and fixed in 10% neutral buffered formalin (NBF) for no longer than 24 hours before processing for morphometric analysis.
  • NBF neutral buffered formalin
  • FIG. 7 shows pancreatic islet size distribution in female C57BL/6J mice at 10 days of peptide treatment.
  • islet size expressed as Log [ ⁇ m 2 ]
  • islet size ranging from 4.9 to 2.3
  • the numbers increased significantly in the mice treated with Peptide 7 (p ⁇ 0.05 or 0.01 versus the naive/control group)( FIG. 7 ).
  • the increase in the Peptide 1 treated mice was only observed in islet size of 2.1 (p ⁇ 0.05 versus the naive/control group).
  • This example describes the effect of peptides on glucose-stimulated insulin secretion (GSIS).
  • pancreases were procured from male adult Sprague-Dawley (SD) rats. After 7 days acclimation, the animals were sacrificed by cervical dislocation and the entire pancreas was removed and digested with collagenase to isolate islets. After digestion, islets were maintained at 37° C.
  • Cultured islets were rinsed in Krebs-Ringer bicarbonate buffer (KRB), pH 7.4, previously gassed with a mixture of CO 2 /O 2 (5/95%), and pre-incubated in 1.0 ml of KRB containing 0.5% (w/v) BSA and 1.5 mM glucose at 37° C. for 45 min. After this period, groups of 5 islets were incubated in 0.6 ml KRB with the addition of 1.5 or 12.0 mM glucose, with or without the addition of peptides for 60 min. At the end of the incubation period, aliquots of the medium were collected for insulin quantitation.
  • KRB Krebs-Ringer bicarbonate buffer
  • FIG. 8 shows the increase of glucose-stimulated insulin secretion of islets with or without the co-incubation of selected peptides (10 ⁇ g/mL), Peptide 12, Peptide 16 and Peptide 1.
  • Co-incubation with 100 nM Glucagon like peptide-1 (GLP-1) was included as a positive control.
  • GLP-1 Glucagon like peptide-1
  • pancreatic islets cultured with peptides GLP-1, Peptide 12 and Peptide 16 released significantly more insulin than those cultured without the addition of peptides.
  • INGAP-PP analogs Peptide 12 and Peptide 16 showed 2-3 fold higher stimulation of insulin secretion than GLP-1.
  • no stimulation was observed with the addition of INGAP-PP (Peptide 1) ( FIG. 8 ).
  • This example describes in vivo pharmacokinetic (PK) properties of peptides in rat and mouse.
  • mice After 7 days acclimation, male Sprague-Dawley (SD) rats weighing 210-250 g, or male C57BL/6 mice, weighing 19-24 g, in good health were used in the study.
  • Peptide 1, Peptide 12 and Peptide 16 were dissolved in sterile normal saline and then they were injected via subcutaneous (sc) bolus or intravenous (iv) bolus at the dose level of 25 mg/kg.
  • sc subcutaneous
  • iv intravenous
  • Blood samples (approximately 400 ⁇ L) were collected and placed into tubes containing EDTA-K2 and centrifuged at 8000 rpm for 6 minutes at 4° C. to separate plasma from the samples. The resulting plasma was stored frozen at ⁇ 80° C. until being analyzed.
  • Plasma concentrations of peptides were determined using tandem mass spectrometry (LC-MS/MS) analysis.
  • the abbreviation AUC (0-t) represents area under the curve from the time of dosing to the time of the last observation, the AUC (0- ⁇ ) represents area under the curve from the time of dosing to infinity, and the C max represents maximum concentration detected.
  • the peptide analogs Peptide 12 and Peptide 16 showed marked improved PK properties evidenced by the significant increase in the area under the plasma concentration-time curves (AUC) and the maximum concentration (Cmax) in mouse and rat.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Diabetes (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Endocrinology (AREA)
  • Cell Biology (AREA)
  • Obesity (AREA)
  • Emergency Medicine (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
US14/775,568 2013-03-15 2013-03-15 Compositions and methods of using islet neogenesis peptides and analogs thereof Abandoned US20160039877A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2013/072771 WO2014139182A1 (en) 2013-03-15 2013-03-15 Compositions and methods of using islet neogenesis peptides and analogs thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2013/072771 A-371-Of-International WO2014139182A1 (en) 2013-03-15 2013-03-15 Compositions and methods of using islet neogenesis peptides and analogs thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/992,039 Continuation US10899815B2 (en) 2013-03-15 2018-05-29 Compositions and methods of using islet neogenesis peptides and analogs thereof

Publications (1)

Publication Number Publication Date
US20160039877A1 true US20160039877A1 (en) 2016-02-11

Family

ID=51535856

Family Applications (7)

Application Number Title Priority Date Filing Date
US14/775,568 Abandoned US20160039877A1 (en) 2013-03-15 2013-03-15 Compositions and methods of using islet neogenesis peptides and analogs thereof
US14/417,110 Active US9388215B2 (en) 2013-03-15 2014-03-14 Compositions and methods of using islet neogenesis peptides and analogs thereof
US15/177,206 Active US9738695B2 (en) 2013-03-15 2016-06-08 Compositions and methods of using islet neogenesis peptides and analogs thereof
US15/654,516 Abandoned US20180009861A1 (en) 2013-03-15 2017-07-19 Compositions and methods of using islet neogenesis peptides and analogs thereof
US15/992,039 Active US10899815B2 (en) 2013-03-15 2018-05-29 Compositions and methods of using islet neogenesis peptides and analogs thereof
US16/658,218 Abandoned US20200347106A1 (en) 2013-03-15 2019-10-21 Compositions and methods of using islet neogenesis peptides and analogs thereof
US17/747,788 Pending US20220348618A1 (en) 2013-03-15 2022-05-18 Compositions and methods of using islet neogenesis peptides and analogs thereof

Family Applications After (6)

Application Number Title Priority Date Filing Date
US14/417,110 Active US9388215B2 (en) 2013-03-15 2014-03-14 Compositions and methods of using islet neogenesis peptides and analogs thereof
US15/177,206 Active US9738695B2 (en) 2013-03-15 2016-06-08 Compositions and methods of using islet neogenesis peptides and analogs thereof
US15/654,516 Abandoned US20180009861A1 (en) 2013-03-15 2017-07-19 Compositions and methods of using islet neogenesis peptides and analogs thereof
US15/992,039 Active US10899815B2 (en) 2013-03-15 2018-05-29 Compositions and methods of using islet neogenesis peptides and analogs thereof
US16/658,218 Abandoned US20200347106A1 (en) 2013-03-15 2019-10-21 Compositions and methods of using islet neogenesis peptides and analogs thereof
US17/747,788 Pending US20220348618A1 (en) 2013-03-15 2022-05-18 Compositions and methods of using islet neogenesis peptides and analogs thereof

Country Status (14)

Country Link
US (7) US20160039877A1 (ru)
EP (2) EP2970385B1 (ru)
JP (2) JP6674258B2 (ru)
KR (1) KR102244349B1 (ru)
AU (3) AU2014231444B2 (ru)
BR (1) BR112015022469A2 (ru)
CA (1) CA2906240C (ru)
ES (1) ES2703110T3 (ru)
IL (1) IL241383B (ru)
MX (2) MX2015012038A (ru)
NZ (1) NZ711958A (ru)
RU (1) RU2685958C2 (ru)
SG (1) SG11201507404RA (ru)
WO (2) WO2014139182A1 (ru)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4560952B2 (ja) * 1999-12-03 2010-10-13 株式会社セガ ゲーム装置及びゲームシステム
CA2901404A1 (en) * 2013-02-15 2014-08-21 The Royal Institution For The Advancement Of Learning/Mcgill University Modified ingap peptides for treating diabetes
US10137088B2 (en) 2016-02-10 2018-11-27 Pfizer Inc. Therapeutic nanoparticles having EGFR ligands and methods of making and using same
CN108616978B (zh) * 2016-12-14 2022-04-15 中兴通讯股份有限公司 一种网络功能实体进行无状态处理的方法及装置
KR102006890B1 (ko) * 2016-12-27 2019-08-05 한양대학교 산학협력단 췌장 소도 세포 표적 펩타이드 및 이의 용도
BR112019023511A2 (pt) * 2017-05-11 2020-05-19 Shenzhen Hightide Biopharmaceutical Ltd uso de compostos peptídeos no tratamento de pancreatite aguda
US20190142901A1 (en) * 2017-11-16 2019-05-16 Claresa Levetan Compositions and methods for treating or preventing type 1 diabetes using a biologic response modifier in combination with one or more islet or beta cell regeneration or replacement therapies
CN114644683B (zh) * 2018-09-18 2023-09-12 广州领晟医疗科技有限公司 促进肝细胞增殖和/或抑制肝细胞凋亡的多肽及其用途
JP2023524061A (ja) * 2020-05-01 2023-06-08 ザ・ブロード・インスティテュート・インコーポレイテッド 操作された中枢神経系用組成物
WO2023143446A1 (en) * 2022-01-25 2023-08-03 Shenzhen Hightide Biopharmaceutical Ltd. Compositions and methods for treating cytokine storm

Family Cites Families (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2661187B1 (fr) 1990-04-20 1994-08-05 Inst Nat Sante Rech Med Proteine associee a la pancreatite aiguue. moyens pour le diagnostic de la pancreatite aiguue.
FR2700011B1 (fr) 1992-12-24 1995-02-24 Inst Nat Sante Rech Med Détection de la mucoviscidose ou d'une mutation du gêne CFTR au moyen d'un dosage de la PAP.
ATE304604T1 (de) 1993-06-24 2005-09-15 Frank L Graham Adenovirus vektoren für gentherapie
DE69434594T2 (de) 1993-10-25 2006-09-21 Canji, Inc., San Diego Rekombinante adenoviren-vektor und verfahren zur verwendung
US7252989B1 (en) 1994-04-04 2007-08-07 Board Of Regents, The University Of Texas System Adenovirus supervector system
US5834590A (en) 1995-02-22 1998-11-10 Eastern Virginia Medical School Of The Medical College Of Hampton Roads Ingap protein involved in pancreatic islet neogenesis
US5688489A (en) 1995-09-15 1997-11-18 Resolution Pharmaceuticals, Inc. Non-receptor mediated imaging agents
US5804421A (en) 1996-10-30 1998-09-08 Eastern Virginia Medical School Of The Medical College Fo Hampton Roads High level of expression of ingap in bacterial and euraryotic cells
US5935813A (en) 1997-03-20 1999-08-10 Incyte Pharmaceuticals, Inc. Human pancreatitis-associated protein
US20020052308A1 (en) 1999-03-12 2002-05-02 Rosen Craig A. Nucleic acids, proteins and antibodies
AU3395900A (en) 1999-03-12 2000-10-04 Human Genome Sciences, Inc. Human lung cancer associated gene sequences and polypeptides
US20040018623A1 (en) 1999-10-29 2004-01-29 Lawrence Rosenberg Medium for preparing dedifferentiated cells
US20020009730A1 (en) 1999-11-17 2002-01-24 Alex Chenchik Human stress array
CA2343602A1 (en) 2000-04-18 2001-10-18 Genset Est's and encoded human proteins
US20070037165A1 (en) 2000-09-08 2007-02-15 Applera Corporation Polymorphisms in known genes associated with human disease, methods of detection and uses thereof
US6812339B1 (en) 2000-09-08 2004-11-02 Applera Corporation Polymorphisms in known genes associated with human disease, methods of detection and uses thereof
US6824822B2 (en) 2001-08-31 2004-11-30 Alkermes Controlled Therapeutics Inc. Ii Residual solvent extraction method and microparticles produced thereby
DE10056802B4 (de) 2000-11-14 2005-06-16 Epigenomics Ag Verfahren zur Detektion von Methylierungszuständen zur toxikologischen Diagnostik
ATE431851T1 (de) 2000-12-13 2009-06-15 Anaphore Inc Methode zur identifikation und isolation von peptide die andere moleküle binden aus kombinatorische proteinbibliotheken mit einer c- typ lectin-ähnlichen domäne als gerüststruktur
AU2002246808A1 (en) 2000-12-19 2002-08-06 Curagen Corporation Human nucleic acids and polypeptides and methods of use thereof
US20040018970A1 (en) 2000-12-19 2004-01-29 Shimkets Richard A. Novel nucleic acids and polypeptides and methods of use thereof
US6986994B2 (en) 2001-01-09 2006-01-17 Gmp Endotherapeutics, Inc. INGAP displacement assays
WO2002070551A2 (en) 2001-03-01 2002-09-12 Mcgill University Neuritogenic compound and uses thereof
EP1478772A2 (en) 2001-08-14 2004-11-24 The General Hospital Corporation Nucleic acid and amino acid sequences involved in pain
MXPA04001804A (es) 2001-08-30 2005-03-07 Biorexis Pharmaceutical Corp Proteinas de fusion de transferrina modificada.
US20040132644A1 (en) * 2001-10-16 2004-07-08 The Procter & Gamble Company Composition and method for treating diabetes
CA2842429A1 (en) 2001-10-19 2003-05-01 Genentech, Inc. Compositions and methods for the diagnosis and treatment of inflammatory bowel disorders
US20060194265A1 (en) 2001-10-23 2006-08-31 Morris David W Novel therapeutic targets in cancer
WO2003059934A2 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
US7662768B2 (en) 2002-01-11 2010-02-16 Mcgill University Transdifferentiation of pancreatic acinar cells
US7166439B2 (en) 2002-03-01 2007-01-23 Gmp Endotherapeutics, Inc. Assay for anti-INGAP antibodies
US20070015271A1 (en) 2002-04-04 2007-01-18 Rosen Craig A Human secreted proteins
US20040018522A1 (en) 2002-05-09 2004-01-29 Brigham And Women's Hospital, Inc. Identification of dysregulated genes in patients with multiple sclerosis
AU2003241388A1 (en) 2002-05-09 2003-11-11 Medtronic Minimed, Inc. Immunoprotective methods for beta cell neogenesis
US20060105333A1 (en) 2002-07-10 2006-05-18 Oncotherapy Science, Inc. Method for diagnosis of intestinal-type gastric tumors
JP2004065120A (ja) 2002-08-07 2004-03-04 Sumitomo Pharmaceut Co Ltd 炎症性腸疾患の疾患マーカー及びその利用
ATE452650T1 (de) 2002-08-30 2010-01-15 Biorexis Pharmaceutical Corp Transferrin-fusionsproteinbibliotheken
US20060009516A1 (en) 2002-10-24 2006-01-12 Mcgill University Use of ingap for reversing diabetes
AU2003294828A1 (en) 2002-12-17 2004-07-09 Sinogenomax Co. Ltd. Chinese National Human Genomecenter Specific markers for pancreatic cancer
US7510708B2 (en) 2003-04-14 2009-03-31 Washington University Disruption of the REG pathway
EP1488798A1 (en) * 2003-06-18 2004-12-22 Institut National De La Sante Et De La Recherche Medicale (Inserm) HIP/PAP polypeptide composition for use in liver regeneration and for the prevention of liver failure
WO2005113812A2 (en) 2004-04-23 2005-12-01 Invitrogen Corporation Collections of matched biological reagents and methods for identifying matched reagents
US20050277593A1 (en) 2004-05-24 2005-12-15 Washington University Therapeutic uses of Reg protein
CN1964987A (zh) 2004-06-04 2007-05-16 惠氏公司 Regⅲ蛋白的抑制剂作为哮喘的治疗剂
KR100639677B1 (ko) 2004-11-08 2006-10-30 삼성전자주식회사 위상 및 지연 동기 루프와 이를 구비한 반도체 메모리 장치
KR100664586B1 (ko) 2004-12-22 2007-01-04 김현기 인간 원암 유전자, 이에 의해 코드되는 단백질, 이를포함하는 발현벡터 및 이 벡터로 형질 전환된 세포
US20090142338A1 (en) * 2005-03-04 2009-06-04 Curedm, Inc. Methods and Compositions for Treating Type 1 and Type 2 Diabetes Mellitus and Related Conditions
US20060275794A1 (en) 2005-03-07 2006-12-07 Invitrogen Corporation Collections of matched biological reagents and methods for identifying matched reagents
US7393919B2 (en) * 2005-05-25 2008-07-01 Cure Dm, Inc. Peptides, derivatives and analogs thereof, and methods of using same
PL3524261T3 (pl) 2005-08-19 2024-04-15 Amylin Pharmaceuticals, Llc Eksendyna do stosowania w leczeniu cukrzycy i zmniejszania masy ciała
US7807459B2 (en) 2005-09-27 2010-10-05 Reneuron, Inc. EphA4-positive human adult pancreatic endocrine progenitor cells
US20080039393A1 (en) 2005-11-09 2008-02-14 Desmond Mascarenhas Metal-binding therapeutic peptides
CA2629113A1 (en) 2005-11-09 2007-05-18 Ontherix, Inc. Metal-binding therapeutic peptides
WO2007071437A2 (en) 2005-12-22 2007-06-28 Ares Trading S.A. Compositions and methods for treating inflammatory disorders
US20090269313A1 (en) 2006-07-19 2009-10-29 Diakine Therapeutics, Inc. Encapsulation system
KR20060089873A (ko) 2006-07-25 2006-08-09 김현기 인간 원암 유전자, 이에 의해 코드되는 단백질, 이를포함하는 발현벡터 및 이 벡터로 형질 전환된 세포
AU2007284651B2 (en) 2006-08-09 2014-03-20 Institute For Systems Biology Organ-specific proteins and methods of their use
WO2008085601A2 (en) 2006-11-02 2008-07-17 Genizon Biosciences Inc. Genemap of the human genes associated with asthma disease
US8785400B2 (en) 2006-11-22 2014-07-22 Curedm Group Holdings, Llc Methods and compositions relating to islet cell neogenesis
WO2008079406A2 (en) 2006-12-19 2008-07-03 Genentech, Inc. Gene expression markers for inflammatory bowel disease
FR2910311B1 (fr) 2006-12-20 2009-02-13 Oreal Composition comprenant un compose silicone et un organosilane particulier
US7923014B2 (en) 2007-02-12 2011-04-12 The Board Of Regents Of The University Of Texas System Expression and purification of HIP/PAP and uses therefor
WO2008118948A1 (en) * 2007-03-26 2008-10-02 Atherogenics, Inc. Methods and compositions of derivatives of probucol for the treatment of diabetes
US20080280985A1 (en) 2007-03-27 2008-11-13 Scott Robert A D Methods and Compositions Using Certain Phenolic Derivatives for the Treatment of Diabetes
US8816047B2 (en) * 2007-08-30 2014-08-26 Cure DM Group Holdings, LLC Compositions and methods of using proislet peptides and analogs thereof
WO2009049222A1 (en) * 2007-10-12 2009-04-16 Curedm, Inc. Compositions and methods of using the human proislet peptide receptor
SG185972A1 (en) 2007-11-07 2012-12-28 Genentech Inc Compositions and methods for treatment of microbial disorders
MX2010005893A (es) 2007-11-29 2011-03-04 Genentech Inc Star Marcadores de expresion genica para enfermedad inflamatoria de intestino.
US20100004213A1 (en) 2007-11-29 2010-01-07 Abbas Alexander R Gene expression markers for inflammatory bowel disease
EP2080812A1 (en) 2008-01-18 2009-07-22 Transmedi SA Compositions and methods of detecting post-stop peptides
WO2009097155A1 (en) 2008-01-31 2009-08-06 Diakine Therapeutics, Inc. Compositions and methods for treating diabetes using lisofylline and islet neogenesis associated peptide
AU2009226246A1 (en) 2008-03-21 2009-09-24 Podiceps B.V. Diagnostic of pre-symptomatic metabolic syndrome
US8680263B2 (en) 2008-09-19 2014-03-25 Nektar Therapeutics Carbohydrate-based drug delivery polymers and conjugates thereof
JP2010085375A (ja) 2008-10-02 2010-04-15 Nationa Hospital Organization 細胞増殖を伴う糖尿病合併症の検査のための方法、組成物およびキット
WO2010062663A1 (en) 2008-11-03 2010-06-03 Schering Corporation Inflammatory bowel disease biomarkers and related methods of treatment
US8012928B2 (en) 2008-12-19 2011-09-06 The Research Foundation Of State University Of New York Truncated PAP2 and methods of making and using same
DK2393828T3 (en) 2009-02-03 2017-01-23 Amunix Operating Inc Extended recombinant polypeptides and compositions comprising same
EP2260857A1 (en) 2009-06-11 2010-12-15 Alfact Innovation Novel applications of HIP/PAP or derivatives thereof
ES2356217B1 (es) 2009-08-04 2012-02-13 Universidad De Salamanca Método para la detección de daño renal.
EP2486132A1 (en) 2009-10-09 2012-08-15 Anaphore, Inc. Combinatorial libraries based on c-type lectin domain
CA2777162A1 (en) 2009-10-09 2011-04-14 Anaphore, Inc. Polypeptides that bind il-23r
US20110312881A1 (en) 2009-12-21 2011-12-22 Amunix, Inc. Bifunctional polypeptide compositions and methods for treatment of metabolic and cardiovascular diseases
ES2374370B1 (es) 2010-08-04 2013-01-08 Universidad De Salamanca Método para la detección de daño renal.
CN102827253B (zh) * 2011-06-17 2017-07-21 上海市第一人民医院 一种抑制炎症反应的小分子多肽及其应用
CA2901404A1 (en) * 2013-02-15 2014-08-21 The Royal Institution For The Advancement Of Learning/Mcgill University Modified ingap peptides for treating diabetes

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Huszarik et al. ("Adjuvant Immunotherapy increased B cell regenerative factor regeneration gene 2 in the pancreas of diabetic mice; J Immunol; 2010, 185) *
Thermo (Thermo Scientific, 2004). *

Also Published As

Publication number Publication date
JP2016512205A (ja) 2016-04-25
AU2020280993B2 (en) 2022-04-21
BR112015022469A2 (pt) 2017-07-18
EP3473642A2 (en) 2019-04-24
IL241383B (en) 2021-03-25
RU2015143655A (ru) 2017-04-26
CA2906240A1 (en) 2014-09-18
EP2970385B1 (en) 2018-10-03
RU2015143655A3 (ru) 2018-02-28
AU2020280993A1 (en) 2021-01-07
JP2020062028A (ja) 2020-04-23
EP2970385A1 (en) 2016-01-20
US20220348618A1 (en) 2022-11-03
RU2685958C2 (ru) 2019-04-23
AU2018241212A1 (en) 2018-11-29
US20170002049A1 (en) 2017-01-05
EP3473642A3 (en) 2019-07-10
US10899815B2 (en) 2021-01-26
CA2906240C (en) 2022-10-18
WO2014139182A1 (en) 2014-09-18
WO2014139472A1 (en) 2014-09-18
US9388215B2 (en) 2016-07-12
SG11201507404RA (en) 2015-10-29
EP3473642B1 (en) 2022-04-13
US20180009861A1 (en) 2018-01-11
NZ711958A (en) 2020-07-31
JP7075918B2 (ja) 2022-05-26
EP2970385A4 (en) 2016-12-07
JP6674258B2 (ja) 2020-04-01
AU2014231444A1 (en) 2015-10-01
ES2703110T3 (es) 2019-03-07
KR20160012992A (ko) 2016-02-03
MX2015012038A (es) 2016-06-02
US20190023755A1 (en) 2019-01-24
AU2014231444B2 (en) 2018-07-19
US20150203538A1 (en) 2015-07-23
IL241383A0 (en) 2015-11-30
US9738695B2 (en) 2017-08-22
MX2019009886A (es) 2019-10-22
AU2018241212B2 (en) 2020-09-17
KR102244349B1 (ko) 2021-04-23
US20200347106A1 (en) 2020-11-05

Similar Documents

Publication Publication Date Title
US10899815B2 (en) Compositions and methods of using islet neogenesis peptides and analogs thereof
EP3157949B1 (en) Exendin-4 derivatives as selective glucagon receptor agonists
CN104045698B (zh) 一种多肽、多肽衍生物、多肽的可药用盐及药物组合物
CN104045703B (zh) 一种多肽、多肽衍生物、多肽的可药用盐及药物组合物
CN104650211A (zh) 一种多肽、多肽衍生物、多肽的可药用盐、药物组合物及其应用
CN104592380A (zh) 一种多肽、多肽衍生物、多肽的可药用盐、药物组合物及其应用
US10829527B2 (en) Conjugates of islet neogenesis peptides and analogs, and methods thereof
CN104045699A (zh) 一种多肽、多肽衍生物、多肽的可药用盐及药物组合物
CN104045700A (zh) 一种多肽、多肽衍生物、多肽的可药用盐及药物组合物
CN104045701A (zh) 一种多肽、多肽衍生物、多肽的可药用盐及药物组合物

Legal Events

Date Code Title Description
AS Assignment

Owner name: SHENZHEN HIGHTIDE BIOPHARMACEUTICAL, LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIU, LIPING;BAI, RU;REEL/FRAME:037371/0979

Effective date: 20151015

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: SHENZHEN HIGHTIDE BIOPHARMACEUTICAL, LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIU, LIPING;BAI, RU;REEL/FRAME:057277/0168

Effective date: 20210824