WO2017039267A1 - Novel insulin analogs and use thereof - Google Patents

Novel insulin analogs and use thereof Download PDF

Info

Publication number
WO2017039267A1
WO2017039267A1 PCT/KR2016/009606 KR2016009606W WO2017039267A1 WO 2017039267 A1 WO2017039267 A1 WO 2017039267A1 KR 2016009606 W KR2016009606 W KR 2016009606W WO 2017039267 A1 WO2017039267 A1 WO 2017039267A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
insulin
insulin analog
chain
tyrosine
Prior art date
Application number
PCT/KR2016/009606
Other languages
French (fr)
Inventor
Jin Young Kim
Euh Lim Oh
Jong Soo Lee
Hyung Kyu Lim
In Young Choi
Se Chang Kwon
Original Assignee
Hanmi Pharm. Co., 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 Hanmi Pharm. Co., Ltd. filed Critical Hanmi Pharm. Co., Ltd.
Priority to JP2018511151A priority Critical patent/JP6976930B2/en
Priority to CA2999823A priority patent/CA2999823A1/en
Priority to CN201680062814.5A priority patent/CN108350056B/en
Priority to AU2016317449A priority patent/AU2016317449B2/en
Priority to EA201890584A priority patent/EA201890584A1/en
Priority to EP16842233.5A priority patent/EP3341404B1/en
Priority to UAA201802993A priority patent/UA125928C2/en
Publication of WO2017039267A1 publication Critical patent/WO2017039267A1/en
Priority to ZA2018/02017A priority patent/ZA201802017B/en
Priority to PH12018500690A priority patent/PH12018500690A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/62Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/12General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by hydrolysis, i.e. solvolysis in general
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/18Ion-exchange chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to a novel insulin analog, and more specifically, to an insulin analog with an improved in vitro effect compared with native insulin, and a use thereof.
  • Insulin is a blood glucose level-controlling hormone secreted by the pancreas, and serves to transport excess glucose in the blood to cells, thereby supplying an energy source and maintaining a normal glucose level.
  • diabetic patients cannot maintain normal insulin functions due to insulin deficiency, insulin resistance, and loss of beta-cell function.
  • diabetic patients cannot utilize the glucose in the blood as an energy source, but show symptoms of hyperglycemia with a high glucose level and excrete the glucose in the urine, which cause of various complications. Accordingly, those diabetic patients who have abnormalities in insulin secretion (type I) or insulin resistance (type II) essentially require insulin treatment, and by insulin administration, they can keep their blood glucose levels normal.
  • Human insulin consists of two polypeptide chains, i.e., the A-chain and the B-chain, which respectively include 21- and 30 amino acids, connected with each other by two disulfide bonds. Since insulin has an extremely short in vivo half-life, as is the case with other protein and peptide hormones, it is unable to show a sustained therapeutic effect, and thus has a problem in that it must be administered continuously and repeatedly to exert its effect. The frequent administration of insulin causes severe pain and discomfort to patients, and thus there is a need to improve the administration from the aspects of patient compliance, safety, and convenience.
  • Insulin is known to remove blood glucose by binding to insulin receptors and the effect of insulin can be controlled by altering the sequence of native insulin.
  • the in vivo effect of insulin can be controlled by substitution of amino acid(s) of insulin with different amino acid(s) or by deletion of specific amino acid(s) of insulin. Since insulin derivatives with high activity can exert effects equivalent to or better than those of native insulin, even in a small amount, they may thus be very desirable from the therapeutic point of view.
  • amino acid substitutions in the A-chain and/or the B-chain contained in insulin have been broadly studied from the aspect of a pharmacokinetic effect of insulin action after subcutaneous injection.
  • the present inventors have intensively studied to improve the effect of insulin action, and as a result, they have discovered that the insulin analogs with modification(s) in particular amino acid residue(s) in the A-chain and/or the B-chain of insulin exhibit a markedly improved in vitro effect compared to that of native insulin, and that they can thus be effectively used for treating diabetes, thereby completing the present invention.
  • An object of the present invention is to provide a novel insulin analog, and specifically an insulin analog with an improved in vitro effect compared with that of native insulin.
  • Another object of the present invention is to provide a pharmaceutical composition for treating diabetes containing the insulin analog as an active ingredient.
  • a further object of the present invention is to provide a method for treating diabetes including administering the insulin analog or a pharmaceutical composition containing the insulin analog as an active ingredient to a subject in need thereof.
  • the present invention provides an insulin analog which includes the A-chain of SEQ ID NO: 3 represented by the following General Formula 1 and the B-chain of SEQ ID NO: 4 represented by the following General Formula 2.
  • Xaa1 is alanine, glycine, glutamine, histidine, glutamic acid, or asparagine;
  • Xaa2 is alanine, glutamic acid, glutamine, histidine, or asparagine;
  • Xaa3 is alanine, serine, glutamine, glutamic acid, histidine, or asparagine;
  • Xaa4 is alanine, tyrosine, glutamic acid, histidine, lysine, aspartic acid, or asparagine;
  • Xaa5 is alanine, leucine, tyrosine, histidine, glutamic acid, or asparagine;
  • Xaa6 is alanine, tyrosine, serine, glutamic acid, histidine, or asparagine;
  • Xaa7 is asparagine, glycine, histidine, or alanine.
  • Xaa8 is tyrosine, glutamic acid, or aspartic acid, or is absent;
  • Xaa9 is phenylalanine, or is absent
  • Xaa10 is threonine, or is absent
  • Xaa11 is proline, glutamic acid, or aspartic acid, or is absent;
  • the insulin analog is characterized in that it includes an A-chain of General Formula 1 and a B-chain, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, and Xaa10 is threonine.
  • the insulin analog is characterized in that it includes an A-chain of General Formula 1 and a B-chain, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, and Xaa10 is absent.
  • the insulin analog is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline.
  • the insulin analog is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is asparagine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline.
  • the insulin analog is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline.
  • the insulin analog is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is alanine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is glutamic acid, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline.
  • the insulin analog according to the present invention is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa4 is glutamic acid, and, in the B-chain of SEQ ID NO: 4, Xaa9 is phenylalanine; or
  • Xaa4 is asparagine
  • Xaa9 is phenylalanine
  • Xaa4 is glutamic acid, and, in the B-chain of SEQ ID NO: 4, Xaa9 is absent; or
  • Xaa4 is alanine
  • Xaa8 is glutamic acid and Xaa9 is absent
  • the insulin analog according to the present invention includes an amino acid sequence represented by SEQ ID NO: 16, 18, 20, or 22.
  • the present invention provides a nucleic acid encoding the insulin analog.
  • the nucleic acid according to the present invention includes nucleotide sequences selected from the group consisting of SEQ ID NOS: 15, 17, 19, and 21.
  • the present invention provides a recombinant expression vector including the nucleic acid.
  • the present invention provides a transformant which is transformed with the recombinant expression vector.
  • the present invention provides a method for preparing the insulin analog including:
  • the present invention provides a pharmaceutical composition for treating diabetes containing the insulin analog as an active ingredient and a pharmaceutically acceptable carrier.
  • the insulin analogs according to the present invention exhibit a significantly improved in vitro effect compared with that of native insulin, and thus the administration of the insulin analogs is expected to provide sufficient treatment even in a small amount, and can thus be effectively used for treating diabetes.
  • FIG. 1 shows a result of the purity of insulin analogs according to the present invention analyzed by protein electrophoresis, and representatively, a result of the insulin analog 1 (Lane 1: size marker, and Lane 2: insulin analog 1).
  • FIG. 2 shows a result of the purity of insulin analogs according to the present invention analyzed by reversed phase chromatography and size exclusion chromatography, and representatively, a result of the insulin analog 1.
  • FIG. 3 shows a result of peptide mapping of the analogs according to the present invention, and representatively, a result of the insulin analog 1, wherein USP-insulin indicates native insulin used as control.
  • the present invention relates to a novel insulin, and specifically an insulin analog with an improved in vitro effect compared with that of native insulin.
  • insulin analog refers to a modified analog of native insulin prepared via modification of a part of the amino acid(s) of native insulin in the form of insertion, deletion, or substitution, and in particular, it includes various insulin analogs of native insulin with an improved in vitro effect compared with that of native insulin.
  • Native insulin is a hormone secreted by the pancreas and generally plays a role in promoting intracellular glucose absorption and inhibiting fat breakdown, thereby controlling in vivo blood glucose levels.
  • Insulin is generated from the processing of its precursor, proinsulin, which does not have the function of controlling blood glucose levels.
  • Insulin is composed of two polypeptide chains, i.e., the A-chain and the B-chain, which include 21 and 30 amino acids, respectively, and are interlinked by a disulfide bridge.
  • Each of the A-chain and the B-chain may include the amino acid sequences represented by SEQ ID NO: 1 and SEQ ID NO: 2 shown below.
  • the insulin according to the present invention refers to insulin analogs prepared by genetic recombination technology, but the insulin is not limited thereto and includes all insulin with an improved in vitro effect compared with that of native insulin.
  • the insulin of the present invention includes inverted insulin, insulin variants, insulin fragments, etc.
  • the insulin can be prepared not only by a recombinant method but also by a solid phase synthesis, and the preparation method is not limited thereto.
  • insulin analogs being peptides having an in vivo blood glucose level-controlling capability equivalent or corresponding to that of native insulin, include all of insulin agonists, insulin derivatives, insulin fragments, insulin variants, etc.
  • insulin agonist refers to a material which can bind to an in vivo receptor of insulin regardless of the structure of insulin and thereby exhibit a biological activity equivalent to that of insulin.
  • insulin derivative may refer to a peptide which has a homology to each of the amino acid sequences of the A-chain and the B-chain of native insulin and is in the form having an in vivo blood glucose level-controlling capability, where a part of the groups in an amino acid residue is modified by chemical substitution (e.g., alpha-methylation, alpha-hydroxylation), deletion (e.g., deamination), or modification (e.g., N-methylation).
  • chemical substitution e.g., alpha-methylation, alpha-hydroxylation
  • deletion e.g., deamination
  • modification e.g., N-methylation
  • insulin derivative may refer to a peptide mimic and a low or high molecular weight compound which can control in vivo blood glucose levels by binding to an insulin receptor, although there is no sequence homology to the amino acid sequence of native insulin.
  • insulin fragment refers to a form of insulin in which at least one amino acid is inserted or deleted, and the amino acid inserted may be one that is not present in nature (e.g., D-type amino acid), and the insulin fragment has an in vivo blood glucose level-controlling capability.
  • insulin variant refers to a peptide which has a difference in at least one amino acid sequence from that of insulin, and the peptide also has the in vivo blood glucose level-controlling capability.
  • the methods used in preparing insulin agonists, derivatives, fragments, and variants may be used independently or in combination.
  • those peptides having the in vivo blood glucose level-controlling capability, which have a difference in at least one amino acid sequence, and in which the amino acid residue in the N-terminus is deaminated may be included in the scope of the present invention.
  • the insulin analog according to the present invention exclusively includes any peptide with an improved in vitro effect compared with that of native insulin by introducing substitution, insertion, or deletion of amino acid(s), or a post-translational modification (e.g., methylation, acylation, ubiquitination, and intermolecular covalent bond) in the amino acid sequences (SEQ ID NOS: 1 and 2) of the A-chain and the B-chain of native insulin.
  • substitution or insertion of the amino acid(s) not only the 20 amino acids conventionally observed in human proteins but also atypical or unnatural amino acids may be used.
  • the atypical amino acids may be commercially obtained from Sigma-Aldrich, ChemPep, Genzymepharmaceuticals, etc.
  • the peptides containing these amino acids and typical peptide sequences may be synthesized by or purchased from commercial peptide synthesis companies, such as American Peptide Company, Bachem (USA), and Anygen (Korea).
  • the insulin analogs according to the present invention may be those which include a modification or deletion in particular amino acid residue(s) of the A-chain and the B-chain of native insulin, and preferably, may be those in which particular amino acid residue(s) of the A-chain of native insulin is(are) modified and particular amino acid residue(s) of the B-chain of native insulin is(are) modified and/or deleted.
  • the insulin analogs of the present invention may be an analog in which the 14 th amino acid residue, tyrosine, in the amino acid sequence of the A-chain represented by SEQ ID NO: 1 is substituted with glutamic acid, asparagine, or alanine, or an analog in which the 16 th amino acid residue, tyrosine, is substituted with glutamic acid and/or the 25 th amino acid residue, phenylalanine, in the amino acid sequence of the B-chain represented by SEQ ID NO: 2 is deleted; or may include all of these.
  • the insulin analogs of the present invention may be those which include the A-chain of SEQ ID NO: 3 represented by the following General Formula 1 and the B-chain of SEQ ID NO: 4 represented by the following General Formula 2.
  • Xaa1 is alanine, glycine, glutamine, histidine, glutamic acid, or asparagine;
  • Xaa2 is alanine, glutamic acid, glutamine, histidine, or asparagine;
  • Xaa3 is alanine, serine, glutamine, glutamic acid, histidine, or asparagine;
  • Xaa4 is alanine, tyrosine, glutamic acid, histidine, lysine, aspartic acid, or asparagine;
  • Xaa5 is alanine, leucine, tyrosine, histidine, glutamic acid, or asparagine;
  • Xaa6 is alanine, tyrosine, serine, glutamic acid, histidine, or asparagine;
  • Xaa7 is asparagine, glycine, histidine, or alanine.
  • Xaa8 is tyrosine, glutamic acid, or aspartic acid, or is absent;
  • Xaa9 is phenylalanine, or is absent
  • Xaa10 is threonine, or is absent
  • Xaa11 is proline, glutamic acid, or aspartic acid, or is absent;
  • the insulin ananlog may be an insulin analog, wherein, in General Formula 1,
  • Xaa1 is glycine
  • Xaa2 is glutamine
  • Xaa3 is serine
  • Xaa4 is alanine, glutamic acid, or asparagine,
  • Xaa5 is leucine
  • Xaa6 is tyrosine
  • Xaa7 is asparagine
  • Xaa8 is tyrosine or glutamic acid
  • Xaa9 is phenylalanine, or is absent
  • Xaa10 is threonine
  • Xaa11 is proline, glutamic acid, or aspartic acid, or is absent,
  • the insulin ananlog may be an insulin analog, wherein, in General Formula 1,
  • Xaa1 is glycine
  • Xaa2 is glutamine
  • Xaa3 is serine
  • Xaa4 is glutamic acid or asparagine
  • Xaa5 is leucine
  • Xaa6 is tyrosine
  • Xaa7 is asparagine
  • Xaa8 is tyrosine
  • Xaa9 is phenylalanine, or is absent
  • Xaa10 is threonine
  • Xaa11 is proline, glutamic acid, or aspartic acid, or is absent,
  • the insulin ananlog may include an A-chain of General Formula 1 and a B-chain, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
  • the insulin ananlog may include an A-chain of General Formula 1 and a B-chain, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is absent, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
  • the insulin ananlog may be characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
  • the insulin ananlog may be characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is asparagine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
  • the insulin ananlog may be characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
  • the insulin ananlog may be characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is alanine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is glutamic acid, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or absent, but is not limited thereto.
  • the insulin analogs of the present invention may include the following analogs:
  • insulin analog 1 A peptide in which the 14 th amino acid residue in the amino acid sequence of the A-chain represented by SEQ ID NO: 3 is glutamic acid and the 25 th amino acid residue in the amino acid sequence of the B-chain represented by SEQ ID NO: 4 is phenylalanine, having an amino acid sequence represented by SEQ ID NO: 16 which is encoded by a nucleic acid containing a nucleotide sequence represented by SEQ ID NO: 15.
  • insulin analog 2 A peptide in which the 14 th amino acid residue in the amino acid sequence of the A-chain represented by SEQ ID NO: 3 is asparagine and the 25 th amino acid residue in the amino acid sequence of the B-chain represented by SEQ ID NO: 4 is phenylalanine, having an amino acid sequence represented by SEQ ID NO: 18 which is encoded by a nucleic acid containing a nucleotide sequence represented by SEQ ID NO: 17.
  • insulin analog 3 A peptide in which the 14 th amino acid residue in the amino acid sequence of the A-chain represented by SEQ ID NO: 3 is glutamic acid and the 25 th amino acid residue in the amino acid sequence of the B-chain represented by SEQ ID NO: 4 is deleted, having an amino acid sequence represented by SEQ ID NO: 20 which is encoded by a nucleic acid containing a nucleotide sequence represented by SEQ ID NO: 19.
  • insulin analog 4 A peptide in which the 14 th amino acid residue in the amino acid sequence of the A-chain represented by SEQ ID NO: 3 is alanine and the 16 th amino acid residue in the amino acid sequence of the B-chain represented by SEQ ID NO: 4 is glutamic acid, and the 25 th amino acid residue is absent, having an amino acid sequence represented by SEQ ID NO: 22 which is encoded by a nucleic acid containing a nucleotide sequence represented by SEQ ID NO: 21.
  • in vitro effect refers to glucose uptake by an insulin analog, and it is indicated by the measurement result of EC 50 on glucose uptake regarding mouse-derived 3T3-L1 cells differentiated into adipocytes.
  • the insulin analog 1 when the in vitro effect of insulin analogs 1 to 3 was measured, the insulin analog 1 showed a 238.4% increase of glucose uptake, the insulin analog 2 showed a 241.7% increase, and the insulin analog 3 showed a 705% increase compared with that of native insulin, respectively, thereby confirming that the insulin analogs according to the present invention exhibit a remarkable in vitro effect of a 2- to 7-fold increase compared with that of native insulin (Table 1).
  • the present invention provides nucleic acids encoding the above insulin analogs.
  • nucleic acid refers to a deoxyribonucleotide (DNA) or ribonucleotide (RNA) including genomic DNA, cDNA, and RNA being transcribed therefrom, and a nucleotide as the basic constituting unit not only includes natural nucleotides but also includes analogues having modifications in a sugar or base (Scheit, Nucleotide Analogs, John Wiley, New York, 1980; Uhlman and Peyman, Chemical Reviews, 90: 543-584, 1990).
  • the nucleic acid of the present invention may be isolated or prepared using standard technology in molecular biology.
  • the nucleic acid of the present invention may be prepared by PCR amplification using appropriate primer sequences based on the gene sequence of native insulin (NM_000207.2, NCBI), and may be prepared by standard synthesis technology using an automated DNA synthesizer.
  • the nucleic acid of the present invention includes the nucleotide sequences represented by SEQ ID NOS: 15, 17, 19, and 21.
  • the nucleic acid of the present invention not only includes the nucleotide sequences represented by SEQ ID NOS: 15, 17, 19, and 21, but also includes all the sequences which have a sequence homology of at least 70% to the above sequences, preferably at least 80%, more preferably at least 90%, even more preferably at least 95%, and most preferably at least 98%, and the peptide encoded by the above nucleic acid can bind to in vivo receptors of insulin, thereby exhibiting a biological activity substantially the same as that of insulin.
  • the term "homology" refers to a degree of similarity with a given amino acid sequence of a native wild-type protein or a polynucleotide sequence encoding the same, and includes those sequences which have the identity of the above-described percentages or higher to the amino acid sequences or polynucleotide sequences of the present invention.
  • the homology may be determined by comparing the two given sequences by the naked eye or may be determined using a bioinformatic algorithm which enables the analysis of a homology by arranging the subject sequences for comparison. The homology between the two given amino acid sequences may be indicated as a percentage.
  • the useful automated algorithm is available for use in GAP, BESTFIT, FASTA, and TFASTA computer software modules of Wisconsin Genetics Software Package (Genetics Computer Group, Madison, WI, USA).
  • the arrangement algorithm automated in the above modules includes sequence arrangement algorithms by Needleman & Wunsch, Pearson & Lipman, and Smith & Waterman.
  • Other useful algorithms on sequence arrangement and homology determination are automated in software including FASTP, BLAST, BLAST2, PSIBLAST, and CLUSTAL W.
  • the present invention provides a recombinant vector including a nucleic acid encoding the insulin analog.
  • the recombinant vector according to the present invention may be constructed as a vector for conventional cloning or expression, and may be constructed as a vector to use a prokaryotic cell or a eukaryotic cell as a host cell.
  • the term "vector” refers to a recombinant vector capable of expressing a target protein in an appropriate host cell, which is a gene construct including essential regulatory factors operably linked to enable the expression of a nucleic acid insert.
  • the present invention can prepare a recombinant vector which includes a nucleic acid encoding an insulin analog, and the insulin analog of the present invention may be obtained via transformation or transfection of the recombinant vector into a host cell.
  • the nucleic acid encoding the insulin analog is operably linked to a promoter.
  • operably linked refers to a functional connection between a regulatory sequence for nucleic acid expression (e.g., a promoter, a signal sequence, a ribosome-binding site, a transcription termination sequence, etc.) and a different nucleotide sequence, and the regulatory sequence can regulate the transcription and/or translation of the different nucleotide sequence by the same.
  • promoter refers to an untranslated nucleic acid sequence located upstream of a coding region, which includes a polymerase-binding site and has the activity of initiating transcription of a gene located downstream of a promoter into mRNA, i.e., a DNA domain to which polymerase binds and initiates the transcription of a gene, and it is located at the 5' domain of mRNA transcription initiation.
  • the vector of the present invention is a recombinant vector and uses a prokaryotic cell as a host cell
  • a strong promoter e.g., tac promoter, lac promoter, lacUV5 promoter, lpp promoter, pL ⁇ promoter, pR ⁇ promoter, rac5 promoter, amp promoter, recA promoter, SP6 promoter, trp promoter, T7 promoter, etc.
  • a strong promoter e.g., tac promoter, lac promoter, lacUV5 promoter, lpp promoter, pL ⁇ promoter, pR ⁇ promoter, rac5 promoter, amp promoter, recA promoter, SP6 promoter, trp promoter, T7 promoter, etc.
  • the vector to be used in the present invention may be prepared by manipulating the plasmids (e.g., pSC101 , pGV1106 , pACYC177 , ColE1, pKT230 , pME290 , pBR322 , pUC8 /9, pUC6 , pBD9 , pHC79 , pIJ61 , pLAFR1 , pHV14, pGEX series, pET series, pPICZ ⁇ series, pUC19 , etc.), phages (e.g., ⁇ gt4 ⁇ B, ⁇ - Charon , ⁇ z1 , M13 , etc.), or viruses (e.g., SV40, etc.) which are commonly used in the art.
  • the plasmids e.g., pSC101 , pGV1106 , pACYC177 , ColE1, pKT230 , pME290 , p
  • the vector of the present invention is a recombinant vector and uses a eukaryotic cell as a host cell
  • promoters derived from the genomes of mammalian cells e.g., metallothionein promoter
  • promoters derived from the mammalian viruses e.g., adenovirus late promoter, 7.5K promoter of papillomavirus, SV40 promoter, cytomegalovirus promoter, and tk promoter of HSV
  • the vector includes a polyadenylated sequence (e.g., bovine growth hormone terminator and a polyadenylated sequence derived from SV40) as a transcription termination sequence.
  • the recombinant vector of the present invention includes an antibiotic-resistance gene commonly used in the art as a selective marker, and may include, for example, genes having resistance to ampicillin, gentamycin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin, neomycin, and tetracycline.
  • the recombinant vector of the present invention may additionally include a different sequence to make it easy to purify target proteins being collected, i.e., a single-chain insulin analog, proinsulin, or an analog thereof.
  • the sequence to be additionally included may be a tag sequence for protein purification, e.g., glutathione S-transferase (Pharmacia, USA), a maltose-binding protein (NEB, USA), FLAG (IBI, USA), 6-histidine, etc., but the kinds of the sequence necessary for the purification of target proteins are not limited thereto.
  • Fusion proteins expressed by the recombinant vector including the above tag sequence may be purified by affinity chromatography.
  • glutathione S-transferase which is the substrate of the enzyme, may be used, and when 6-histidine tag is used, a desired target protein may be easily collected by a Ni-NTA column.
  • the present invention provides a transformant transformed by a recombinant vector including the nucleic acid encoding the insulin analog.
  • transformation refers to a process of introducing DNA into a host cell and making the DNA replicable therein as a chromosomal factor or by completion of chromosomal integration, which is a phenomenon of artificially causing a genetic change by introducing exogenous DNA into a cell.
  • the method of transformation used in the present invention may be any transformation method, and it may be easily performed according to the conventional method used in the art.
  • Examples of the commonly used transformation method may include a CaCl 2 precipitation method, the Hanahan method with improved efficiency using dimethyl sulfoxide (DMSO) as a reducing agent in the CaCl 2 precipitation method, electroporation, a CaPO 4 precipitation method, a protoplast fusion method, a stirring method using silicon carbide fiber, an agrobacteria-mediated transformation, a transformation using PEG, dextran sulfate-, lipofectamine-, and dry/suppression-mediated transformations, etc.
  • DMSO dimethyl sulfoxide
  • the method for transforming the recombinant vector including a nucleic acid encoding an insulin analog according to the present invention may not be limited to these methods, but any method for transformation or transfection commonly used in the art may be used without limitation.
  • the transformant of the present invention may be obtained by introducing a recombinant vector including the target nucleic acid which encodes an insulin analog into a host cell.
  • a recombinant vector including the target nucleic acid which encodes an insulin analog into a host cell.
  • an appropriate host to be used in the present invention may not be particularly limited as long as it can express the nucleic acid of the present invention.
  • Examples of the appropriate host may include a bacteria belonging to the genus Escherichia such as E.
  • E. coli a bacteria belonging to the genus Bacillus such as Bacillus subtilis , a bacteria belonging to the genus Pseudomonas such as Pseudomonas putida , yeasts such as Pichia pastoris , Saccharomyces cerevisiae , and Schizosaccharomyces pombe , an insect cell such as Spodoptera frugiperda (SF9), and animal cells such as CHO, COS, and BSC.
  • E. coli is used as a host cell.
  • the respective nucleotide sequence encoding the insulin analogs 1 to 3 according to the present invention was amplified via PCR, and the amplified gene fragments were cloned into pET22b vector (Novagen).
  • the pET22b vector was treated with restriction enzymes, Nde I and BamH I, to remove a signal sequence therein, the PCR-amplified products of the insulin analogs were treated with the same restriction enzymes, Nde I and BamH I, and the respective isolated DNA was inserted into the pET22b cloning vector using T4 DNA ligase.
  • the thus-obtained expression vectors were named as pET22b -insulin analogs 1 to 4, respectively.
  • the expression vector pET22b -insulin analogs 1 to 4 respectively encode amino acid sequences represented by SEQ ID NOS: 16, 18, 20, and 22, under the control of T7 promoter, and each of the insulin analogs was expressed in the form of an inclusion body in a host cell, respectively.
  • the recombinant vector pET22b -insulin analogs 1 to 4 including nucleic acids encoding each of the insulin analogs of SEQ ID NOS: 16, 18, 20, and 22 were transformed into E. coli , respectively, and thereby transformants expressing them in the form of an inclusion body were obtained.
  • the present invention provides a method for preparing an insulin analog using the transformants.
  • the present invention provides a method for preparing an insulin analog, including:
  • the medium used in culturing the transformants in the present invention should meet the requirements for host cell cultivation in an appropriate manner.
  • the carbon sources to be contained in the medium for the growth of a host cell may be appropriately selected by the decision of a skilled person in the art according to the transformants prepared thereof, and appropriate cultivation conditions may be selected to control the period and amount of cultivation.
  • sugars and carbohydrates such as glucose, saccharose, lactose, fructose, maltose, starch, and cellulose
  • oils and fats such as soybean oil, sunflower oil, castor oil, and coconut oil
  • fatty acids such as palmitic acid, stearic acid, and linoleic acid
  • alcohols such as glycerol and ethanol
  • organic acids such as acetic acid.
  • nitrogen source to be used may include peptone, yeast extract, meat gravy, malt extract, corn steep liquor, soybean flour, and urea, or inorganic compounds such as ammonium sulfate, ammonium chloride, ammonium phosphate, ammonium carbonate, and ammonium nitrate.
  • the nitrogen source may also be used alone or in combination.
  • Examples of the phosphorous source to be used may include potassium dihydrogen phosphate or dipotassium hydrogen phosphate or a corresponding sodium-containing salt.
  • the culture media may contain a metal salt such as magnesium sulfate or iron sulfate necessary for the growth of the transformant.
  • essential growth materials such as amino acids and vitamins may be used.
  • appropriate precursors for culture media may also be used.
  • the above sources may be appropriately added to a culture during cultivation by a batch culture or continuous culture.
  • the pH of the culture may be appropriately adjusted using a basic compound such as sodium hydroxide, potassium hydroxide, and ammonia, or an acid compound such as phosphoric acid or sulfuric acid.
  • an antifoaming agent such as fatty acid polyglycol ester may be added to prevent foam generation.
  • oxygen or an oxygen-containing gas e.g., air
  • the transformant of the present invention may be cultured at 20°C to 45°C, and preferably, 25°C to 40°C. Additionally, the cultivation is continued until the maximum amount of production of the desired insulin analogs is obtained, and in this regard, the cultivation may normally be continued for 10 hours to 160 hours.
  • the transformant of the present invention can produce insulin analogs when appropriate culture conditions are provided according to host cells, and the peptide-N-glycosidase produced thereof according to the vector constitution and characteristics of a host cell may be secreted within the cytoplasm or into the periplasmic space of the host cell or extracellularly.
  • the proteins expressed within or outside of the host cell may be purified by a conventional method.
  • Examples of the purification method may include salting-out (e.g., ammonium sulfate precipitation, ammonium phosphate precipitation, etc.), solvent precipitation (e.g., protein fraction precipitation using acetone or ethanol, etc.), dialysis, gel filtration, ion exchange, or chromatography such as reversed column chromatography, ultrafiltration, etc., and these methods may be used alone or in combination.
  • salting-out e.g., ammonium sulfate precipitation, ammonium phosphate precipitation, etc.
  • solvent precipitation e.g., protein fraction precipitation using acetone or ethanol, etc.
  • dialysis gel filtration, ion exchange, or chromatography such as reversed column chromatography, ultrafiltration, etc.
  • the transformant of the present invention is characterized in that the insulin analogs 1 to 3 are expressed from the recombinant vector pET22b -insulin analogs 1 to 3 in the form of an inclusion body under the control of T7 promoter. Accordingly, it is preferable that the insulin analogs 1 to 3, which were expressed in the form of an inclusion body, are converted into a soluble form and then isolated and purified.
  • the present invention may further include the following steps for isolating and purifying the insulin analogs expressed in the form of an inclusion body from the transformant:
  • the present invention provides a pharmaceutical composition for treating diabetes containing the above-mentioned insulin analogs.
  • the pharmaceutical composition containing the insulin analogs according to the present invention may contain a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier for oral administration may include a binder, a glidant, a disintegrating agent, an excipient, a solubilizing agent, a dispersing agent, a stabilizing agent, a suspending agent, a coloring agent, a flavoring agent, etc.; for injection formulations, a buffering agent, a preserving agent, an analgesic, an isotonic agent, a stabilizing agent, etc. may be mixed for use; and for topical formulations, a base, an excipient, a lubricant, a preserving agent, etc. may be used.
  • the formulation type of the pharmaceutical composition according to the present invention may be prepared variously by combination with the pharmaceutically acceptable carriers described above.
  • the pharmaceutical composition may be formulated into tablets, troches, capsules, elixirs, suspensions, syrups, wafers, etc.
  • the pharmaceutical composition may be formulated into single-dose ampoules or multidose containers.
  • the pharmaceutical composition may also be formulated into solutions, suspensions, tablets, pills, capsules, and sustained-release formulations.
  • suitable carriers, excipients, and diluents may include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinylpyrrolidone, water, methyl hydroxybenzoate, propyl hydroxybenzoate, talc, magnesium stearate, mineral oil, etc.
  • the pharmaceutical composition of the present invention may further contain a filler, an anti-coagulant, a lubricant, a humectant, a flavoring agent, an emulsifier, a preservative, etc.
  • the present invention provides a method for treating diabetes including administering a pharmaceutical composition containing the insulin analogs of the present invention to a subject in need thereof.
  • the insulin analogs according to the present invention exhibit a significantly improved in vitro effect compared with that of native insulin, and thus it is expected that the administration of a pharmaceutical composition containing the above insulin analogs can be effective for treating diabetes.
  • the term "administration" refers to introduction of a particular material to a patient by an appropriate manner, and the conjugate of the present invention may be administered via any of the common routes as long as the drug can arrive at a target tissue.
  • intraperitoneal, intravenous, intramuscular, subcutaneous, intradermal, oral, topical, intranasal, intrapulmonary, and intrarectal administration may be performed, but the administration route is not limited thereto.
  • active ingredients of a composition for oral administration should be coated or formulated for protection against degradation in the stomach.
  • the present composition may be administered in an injectable form.
  • the pharmaceutical composition may be administered using a certain apparatus capable of transporting the active ingredients into a target cell.
  • the pharmaceutical composition of the present invention may be determined by the types of the drug as an active component as well as by several related factors including the types of diseases to be treated, administration routes, age, sex, and weight of a patient, and severity of the illness. Since the pharmaceutical composition of the present invention has excellent in vivo duration and titer, it can considerably reduce the administration frequency and dose of pharmaceutical drugs of the present invention.
  • primer pairs consisting of a forward primer and a reverse primer for amplifying the insulin analogs introduced with the corresponding modification were synthesized, and PCR was then performed using proinsulin cDNA as a template.
  • the template used was that in which proinsulin cDNA (SC128255, OriGene) (see sequences: BC005255.1 and AAH05255) was cloned into pET22b vector (Novagen), and for smooth recombinant expression of insulin, the nucleotide sequence of SEQ ID NO: 23 (ATG GCA ACA ACA TCA ACA GCA ACT ACG CGT), which encodes the amino acid sequence of Met Ala Thr Thr Ser Thr Ala Thr Thr Arg (SEQ ID NO: 24), was inserted into the cloned proinsulin cDNA as a N-terminal fusion partner.
  • insulin analogs including the amino acid modifications shown in Table 1 were synthesized.
  • Insulin Analog 1 A 14 Tyr ⁇ Glu Insulin Analog 2 A 14 Tyr ⁇ Asn Insulin Analog 3 A 14 Tyr ⁇ Glu + B 25 deletion Insulin Analog 4 A 14 Tyr ⁇ Ala + B 16 Tyr ⁇ Glu, B 25 deletion
  • the insulin analog 1 is an analog which includes a substitution of the 14 th amino acid in the amino acid sequence of the A-chain of native insulin represented by SEQ ID NO: 1, i.e., tyrosine, with glutamic acid;
  • the insulin analog 2 is an analog which includes a substitution of the 14 th amino acid in the amino acid sequence of the A-chain of native insulin represented by SEQ ID NO: 1, i.e., tyrosine, with asparagine;
  • the insulin analog 3 is an analog which includes a substitution of the 14 th amino acid in the amino acid sequence of the A-chain of native insulin represented by SEQ ID NO: 1, i.e., tyrosine, with glutamic acid, and a deletion of the 25 th amino acid in the amino acid sequence of the B-chain of native insulin represented by SEQ ID NO: 2, i.e., phenylalanine, and
  • the insulin analog 4 is an analog which includes a substitution of the 14 th amino acid in the amino acid sequence of the A-chain of native insulin represented by SEQ ID NO: 1, i.e., tyrosine, with alanine, and a substitution of the 16 th amino acid in the amino acid sequence of the B-chain of native insulin represented by SEQ ID NO: 2, i.e., tyrosine, with glutamic acid and a deletion of the 25 th amino acid in the amino acid sequence of the B-chain of native insulin represented by SEQ ID NO: 2, i.e., phenylalanine.
  • the primer pair consisting of SEQ ID NOS: 5 and 6 was designed for the substitution of the 14 th amino acid in the amino acid sequence of the A-chain of native insulin, i.e., tyrosine, with glutamic acid;
  • the primer pair consisting of SEQ ID NOS: 7 and 8 was designed for the substitution of the 14 th amino acid in the amino acid sequence of the A-chain of native insulin, i.e., tyrosine, with asparagine;
  • the primer pair consisting of SEQ ID NOS: 9 and 10 was designed for the deletion of the 25 th amino acid in the amino acid sequence of the B-chain of native insulin, i.e., phenylalanine;
  • the primer pair consisting of SEQ ID NOS: 11 and 12 was designed for the substitution of the 14 th amino acid in the amino acid sequence of the A-chain of native insulin i.e., tyrosine, with alanine;
  • the primer pair consisting of SEQ ID NOS: 13 and 14 was
  • a reaction solution was prepared by mixing 150 ng of template DNA, 1 mL each of 100 pM primers, 5 mL of 2.5 mM dNTP, 10 units of pfx polymerase (Invitrogen, USA), and a 10X buffer solution.
  • the reaction solution was subjected to initial denaturation at 95°C for 30 seconds, followed by 18 repeated cycles of annealing at 95°C for 30 seconds, 55°C for 30 seconds, and 68°C for 6 minutes, and it was finally left at 68°C for 5 minutes.
  • the thus-obtained PCR-amplified products were extracted using a gel extraction kit (Qiagen, Germany) and treated with restriction enzymes, Nde I and BamH I, to prepare insertion fragments.
  • the pET22b vector (Novagen, USA) was then cleaved with the same restriction enzymes and fragments were extracted using the same gel extraction kit.
  • the above insertion fragments were ligated into the thus-prepared vector using T4 ligase to prepare expression vector pET22b -insulin analogs 1 to 4.
  • the expression vectors include nucleic acids encoding the amino acid sequences of the insulin analogs 1 to 4 under the control of T7 promoter, and the vectors can express the insulin analog proteins in the form of an inclusion body in a host cell.
  • the thus-obtained expression vector pET22b -insulin analog 1 according to the present invention includes nucleic acid having a nucleotide sequence represented by SEQ ID NO: 15, which encodes the insulin analog having an amino acid sequence represented by SEQ ID NO: 16;
  • the thus-obtained expression vector pET22b -insulin analog 2 according to the present invention includes a nucleic acid having a nucleotide sequence represented by SEQ ID NO: 17 which encodes the insulin analog having an amino acid sequence represented by SEQ ID NO: 18;
  • the thus-obtained expression vector pET22b -insulin analog 3 according to the present invention includes a nucleic acid having a nucleotide sequence represented by SEQ ID NO: 19, which encodes the insulin analog having an amino acid sequence represented by SEQ ID NO: 20;
  • the thus-obtained expression vector pET22b -insulin analog 4 according to the present invention includes a nucleic acid having a nucleotide sequence represented by SEQ ID NO
  • E . coli BL21-DE3 E. coli B F-dcm ompT hsdS(rB - mB - ) gal ⁇ DE3 (Novagen, USA) was transformed with each of the insulin analog expression vectors prepared in Example 1. Transformation was performed using a method recommended by Novagen, the manufacturer of E. coli BL21- DE3 . Each single colony transformed with the insulin analog expression vectors was collected, inoculated into a 2X Luria Broth (LB) medium containing 50 ⁇ g/mL ampicillin, and cultured at 37°C for 15 hours. The recombinant E.
  • LB 2X Luria Broth
  • recombinant insulin analogs For the expression of recombinant insulin analogs, one vial of each cell stock was dissolved in 500 mL of 2X LB and incubated in a shaking water bath maintained at 37°C for 14 hours to 16 hours. The incubation was stopped when the OD value reached 5.0 or higher, and the culture was used as a seed culture.
  • the seed culture was inoculated into 17 L of a fermentation medium using a 50 L fermenter (MSJ-U2, B.E. MARUBISHI, Japan) and the initial batch fermentation was started. The cultivation was performed at 37°C at a stirring rate of 500 rpm with 20 L/min (1 vvm) of air supply while maintaining the pH at 6.70 with 30% ammonia water.
  • the fermentation was carried out in a fed-batch culture by adding a feeding solution.
  • the growth of bacteria was monitored based on OD values, and when the OD value reached 100 or higher, IPTG at a final concentration of 500 ⁇ M was introduced therein.
  • the cultivation was continued further for about 23 hours to 25 hours after the introduction.
  • the recombinant bacteria was recovered by centrifugation and stored at -80°C until use.
  • Example 3 Isolation and purification of recombinant insulin analogs
  • Example 2 For the isolation and purification of the recombinant insulin analogs expressed in Example 2 from the transformants, cells were disrupted as shown below followed by refolding in order to change the insulin analogs expressed in the form of a water-insoluble inclusion body to a water-soluble form.
  • each cell pellet was resuspended in a 1 L solubilizing buffer solution (50 mM Tris-HCl (pH 9.0), 1 mM EDTA (pH 8.0), 0.2 M NaCl, and 0.5% Triton X-100), and the cells were disrupted using a microfluidizer M-110EH (AC Technology Corp. Model M1475C) at a pressure of 15,000 psi.
  • the disrupted cell lysates were centrifuged at 7,000 rpm and 4°C for 20 minutes and the supernatant was discarded.
  • the resultant was resuspended in 3 L of a washing buffer (0.5% Triton X-100, 50 mM Tris (pH 8.0), 0.2 M NaCl, and 1 mM EDTA). Centrifugation was performed at 7,000 rpm and 4°C for 20 minutes, and the resulting pellet was resuspended in distilled water, followed by centrifugation in the same manner. Each of the resulting pellets was resuspended in 400 mL of a buffer solution (1 M glycine, 3.78 g cysteine-HCl, pH 10.6) and stirred at room temperature for 1 hour.
  • a buffer solution (1 M glycine, 3.78 g cysteine-HCl, pH 10.6
  • Example ⁇ 3-1> The samples in which the refolding was completed in Example ⁇ 3-1> were respectively loaded into a cation exchange column (Source S, GE Healthcare), which was equilibrated with a 20 mM sodium citrate buffer solution (pH 2.0) containing 45% ethanol to be conjugated thereto. Insulin analog proteins were then eluted from the column with a linear concentration gradient from 0% to 100% in 10 column volumes using a 20 mM sodium citrate buffer solution (pH 2.0) which contained 0.5 M potassium chloride and 45% ethanol.
  • a cation exchange column Source S, GE Healthcare
  • Example ⁇ 3-2> Salts were removed from the samples eluted in Example ⁇ 3-2> using a desalting column, followed by replacement of a buffer solution (10 mM Tris-HCl, pH 8.0).
  • the samples were treated with trypsin, which corresponds to a molar ratio of 1000 relative to the protein amount of the sample, and carboxypeptidase B, which corresponds to a molar ratio of 2000 relative to the protein amount of the sample, and stirred at 16°C for 16 hours.
  • the reaction was stopped by lowering the pH to 3.5 using 1 M sodium citrate (pH 2.0).
  • Example ⁇ 3-3> The samples in which the reaction was completed in Example ⁇ 3-3> were respectively reloaded into a cation exchange column (Source S, GE Healthcare) which was equilibrated with a 20 mM sodium citrate buffer solution (pH 2.0) containing 45% ethanol to be conjugated thereto. Insulin analog proteins were then eluted from the column with a linear concentration gradient from 0% to 100% in 10 column volumes using a 20 mM sodium citrate buffer solution (pH 2.0) which contained 0.5 M potassium chloride and 45% ethanol.
  • a cation exchange column Source S, GE Healthcare
  • Example ⁇ 3-4> Salts were removed from the samples eluted in Example ⁇ 3-4> using a desalting column, followed by replacement of a buffer solution (10 mM Tris-HCl, pH 7.5).
  • a buffer solution 10 mM Tris-HCl, pH 7.5
  • the resultants were respectively loaded into an anion exchange column (Source Q, GE Healthcare) equilibrated with a 10 mM Tris buffer solution (pH 7.5) to be conjugated. Insulin analog proteins were then eluted from the column with a linear concentration gradient from 0% to 100% in 10 column volumes using a 10 mM Tris buffer solution (pH 7.5) which contained 0.5 M sodium chloride.
  • the purity of the purified insulin analogs was analyzed via protein electrophoresis (SDS-PAGE) and reversed phase and size exclusion chromatography, and the results are shown in FIG. 1 and FIG. 2, respectively. Additionally, the modifications in amino acids were confirmed by peptide mapping and the analysis of molecular weight of each peak, and the results are shown in FIG. 3.
  • the 3T3-L1 cell line was suspended in a differentiation medium (DMEM containing 10% FBS), inoculated into a 48-well plate at a concentration of 5X10 4 cells/well, and cultured at 37°C for 48 hours.
  • a differentiation medium DMEM containing 10% FBS
  • the differentiation medium was treated with 1 ⁇ g/mL of human insulin (Sigma, Cat. No. I9278), 0.5 mM IBMX (3-isobutyl-1-methylxanthine, Sigma, Cat. No. I5879), and 1 ⁇ M dexamethasone (Sigma, Cat. No. D4902), and the existing medium was removed and the mixture was aliquoted into each well in the amount of 250 ⁇ L/well.
  • the medium was replaced with a differentiation medium to which only 1 ⁇ g/mL of human insulin was added.
  • the induction of differentiation of the 3T3-L1 cell line into adipocytes was then confirmed for a period of 7 to 9 days while replacing the medium with the differentiation medium containing 1 ⁇ g/mL of human insulin at 48 hour intervals.
  • the cells which completed their differentiation into adipocytes were washed once with a serum-free DMEM medium, and then treated with 250 ⁇ L of the serum-free DMEM medium for 4 hours to induce serum depletion therein.
  • Human insulin and insulin analogs were respectively subjected to a 10-fold serial dilution from 5 ⁇ M to 0.005 nM using serum-free DMEM medium to be used as samples.
  • the thus-prepared insulin samples were respectively added into cells in an amount of 250 ⁇ L, and cultured at 37°C for 24 hours in a 5% CO 2 incubator.
  • each culture sample was collected in an amount of 200 ⁇ L, diluted 5-fold using D-PBS, and subjected to the GOPOD analysis (GOPOD Assay Kit, Megazyme, Cat. No. K-GLUC).
  • the concentration of the remaining glucose was calculated based on the absorbance of a glucose standard solution, the EC 50 values on glucose uptake capability of the insulin analogs were respectively calculated, and the results are shown in Table 4 below.
  • the insulin analog 1 showed a 238.4% increase of glucose uptake capability
  • the insulin analog 2 showed a 241.7% increase
  • the insulin analog 3 showed a 705% increase, compared with that of native insulin, respectively.
  • the insulin analogs according to the present invention exhibit a remarkable in vitro effect of a 2- to 7-fold increase compared with that of native insulin, and these results indicate that the insulin analogs can significantly increase their in vivo serum half-life and can thus be provided as stable insulin formulations, thus being effectively used as a therapeutic agent for treating diabetes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Diabetes (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Endocrinology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Analytical Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Emergency Medicine (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to a novel insulin analog, and more specifically, to an insulin analog with an improved in vitro effect compared with native insulin, a nucleic acid encoding the same, an expression vector including the nucleic acid, a transformant introduced with the expression vector, a method of producing the insulin analog from the transformant, a pharmaceutical composition for treating diabetes containing the insulin analog as an active ingredient, and a method for treating diabetes using the insulin analog or the pharmaceutical composition.

Description

NOVEL INSULIN ANALOGS AND USE THEREOF
The present invention relates to a novel insulin analog, and more specifically, to an insulin analog with an improved in vitro effect compared with native insulin, and a use thereof.
Insulin is a blood glucose level-controlling hormone secreted by the pancreas, and serves to transport excess glucose in the blood to cells, thereby supplying an energy source and maintaining a normal glucose level. However, diabetic patients cannot maintain normal insulin functions due to insulin deficiency, insulin resistance, and loss of beta-cell function. As a result, diabetic patients cannot utilize the glucose in the blood as an energy source, but show symptoms of hyperglycemia with a high glucose level and excrete the glucose in the urine, which cause of various complications. Accordingly, those diabetic patients who have abnormalities in insulin secretion (type I) or insulin resistance (type II) essentially require insulin treatment, and by insulin administration, they can keep their blood glucose levels normal.
Human insulin consists of two polypeptide chains, i.e., the A-chain and the B-chain, which respectively include 21- and 30 amino acids, connected with each other by two disulfide bonds. Since insulin has an extremely short in vivo half-life, as is the case with other protein and peptide hormones, it is unable to show a sustained therapeutic effect, and thus has a problem in that it must be administered continuously and repeatedly to exert its effect. The frequent administration of insulin causes severe pain and discomfort to patients, and thus there is a need to improve the administration from the aspects of patient compliance, safety, and convenience.
Accordingly, studies have focused on the development of various protein formulations, chemical conjugates, etc. for improving the therapeutic effects as well as the quality of patients' lives by reducing the frequency of administration through the increase of the in vivo half-life of these protein drugs such as insulin.
Insulin is known to remove blood glucose by binding to insulin receptors and the effect of insulin can be controlled by altering the sequence of native insulin. The in vivo effect of insulin can be controlled by substitution of amino acid(s) of insulin with different amino acid(s) or by deletion of specific amino acid(s) of insulin. Since insulin derivatives with high activity can exert effects equivalent to or better than those of native insulin, even in a small amount, they may thus be very desirable from the therapeutic point of view. In particular, amino acid substitutions in the A-chain and/or the B-chain contained in insulin have been broadly studied from the aspect of a pharmacokinetic effect of insulin action after subcutaneous injection.
Under these circumstances, the present inventors have intensively studied to improve the effect of insulin action, and as a result, they have discovered that the insulin analogs with modification(s) in particular amino acid residue(s) in the A-chain and/or the B-chain of insulin exhibit a markedly improved in vitro effect compared to that of native insulin, and that they can thus be effectively used for treating diabetes, thereby completing the present invention.
An object of the present invention is to provide a novel insulin analog, and specifically an insulin analog with an improved in vitro effect compared with that of native insulin.
Another object of the present invention is to provide a pharmaceutical composition for treating diabetes containing the insulin analog as an active ingredient.
A further object of the present invention is to provide a method for treating diabetes including administering the insulin analog or a pharmaceutical composition containing the insulin analog as an active ingredient to a subject in need thereof.
In order to achieve the above objects, in an aspect, the present invention provides an insulin analog which includes the A-chain of SEQ ID NO: 3 represented by the following General Formula 1 and the B-chain of SEQ ID NO: 4 represented by the following General Formula 2.
General Formula 1
Xaa1-Ile-Val-Glu-Xaa2-Cys-Cys-Thr-Ser-Ile-Cys-Xaa3-Leu-Xaa4-Gln-Xaa5-Glu-Asn-Xaa6-Cys-Xaa7 (SEQ ID NO: 3)
In the above General Formula 1,
Xaa1 is alanine, glycine, glutamine, histidine, glutamic acid, or asparagine;
Xaa2 is alanine, glutamic acid, glutamine, histidine, or asparagine;
Xaa3 is alanine, serine, glutamine, glutamic acid, histidine, or asparagine;
Xaa4 is alanine, tyrosine, glutamic acid, histidine, lysine, aspartic acid, or asparagine;
Xaa5 is alanine, leucine, tyrosine, histidine, glutamic acid, or asparagine;
Xaa6 is alanine, tyrosine, serine, glutamic acid, histidine, or asparagine; and
Xaa7 is asparagine, glycine, histidine, or alanine.
General Formula 2
Phe-Val-Asn-Gln-His-Leu-Cys-Gly-Ser-His-Leu-Val-Glu-Ala-Leu-Xaa8-Leu-Val-Cys-Gly-Glu-Arg-Gly-Phe-Xaa9-Tyr-Xaa10-Xaa11-Lys-Thr (SEQ ID NO: 4)
In the above General Formula 2,
Xaa8 is tyrosine, glutamic acid, or aspartic acid, or is absent;
Xaa9 is phenylalanine, or is absent;
Xaa10 is threonine, or is absent; and
Xaa11 is proline, glutamic acid, or aspartic acid, or is absent;
(with the proviso that the peptides comprising the A-chain of SEQ ID NO: 1 and the B-chain of SEQ ID NO: 2 is excluded).
In a more specific exemplary embodiment, the insulin analog is characterized in that it includes an A-chain of General Formula 1 and a B-chain, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, and Xaa10 is threonine.
The insulin analog is characterized in that it includes an A-chain of General Formula 1 and a B-chain, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, and Xaa10 is absent.
In another exemplary embodiment, the insulin analog is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline.
In still another exemplary embodiment, the insulin analog is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is asparagine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline.
In still another exemplary embodiment, the insulin analog is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline.
In still another exemplary embodiment, the insulin analog is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is alanine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is glutamic acid, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline.
In still another exemplary embodiment, the insulin analog according to the present invention is characterized in that, in the A-chain of SEQ ID NO: 3, Xaa4 is glutamic acid, and, in the B-chain of SEQ ID NO: 4, Xaa9 is phenylalanine; or
in the A-chain of SEQ ID NO: 3, Xaa4 is asparagine, and, in the B-chain of SEQ ID NO: 4, Xaa9 is phenylalanine; or
in the A-chain of SEQ ID NO: 3, Xaa4 is glutamic acid, and, in the B-chain of SEQ ID NO: 4, Xaa9 is absent; or
in the A-chain of SEQ ID NO: 3, Xaa4 is alanine, and, in the B-chain of SEQ ID NO: 4, Xaa8 is glutamic acid and Xaa9 is absent,
but is not limited thereto.
In still another exemplary embodiment, the insulin analog according to the present invention includes an amino acid sequence represented by SEQ ID NO: 16, 18, 20, or 22.
In another aspect, the present invention provides a nucleic acid encoding the insulin analog.
In an exemplary embodiment, the nucleic acid according to the present invention includes nucleotide sequences selected from the group consisting of SEQ ID NOS: 15, 17, 19, and 21.
In still another aspect, the present invention provides a recombinant expression vector including the nucleic acid.
In still another aspect, the present invention provides a transformant which is transformed with the recombinant expression vector.
In still another aspect, the present invention provides a method for preparing the insulin analog including:
a) preparing a recombinant expression vector including a nucleic acid encoding the insulin analog peptide;
b) transforming the recombinant expression vector into a host cell and obtaining a transformant therefrom;
c) culturing the transformant and expressing the insulin analog peptide; and
d) isolating and purifying the expressed insulin analog peptide.
In still another aspect, the present invention provides a pharmaceutical composition for treating diabetes containing the insulin analog as an active ingredient and a pharmaceutically acceptable carrier.
The insulin analogs according to the present invention exhibit a significantly improved in vitro effect compared with that of native insulin, and thus the administration of the insulin analogs is expected to provide sufficient treatment even in a small amount, and can thus be effectively used for treating diabetes.
FIG. 1 shows a result of the purity of insulin analogs according to the present invention analyzed by protein electrophoresis, and representatively, a result of the insulin analog 1 (Lane 1: size marker, and Lane 2: insulin analog 1).
FIG. 2 shows a result of the purity of insulin analogs according to the present invention analyzed by reversed phase chromatography and size exclusion chromatography, and representatively, a result of the insulin analog 1.
FIG. 3 shows a result of peptide mapping of the analogs according to the present invention, and representatively, a result of the insulin analog 1, wherein USP-insulin indicates native insulin used as control.
The present invention will be described in greater detail hereinbelow.
Meanwhile, each of the explanations and exemplary embodiments disclosed herein can be applied to other explanations and exemplary embodiments. That is, all possible combinations of various elements disclosed herein belong to the scope of the present invention. Additionally, the scope of the present invention should not be limited by the specific descriptions provided hereinbelow.
The present invention relates to a novel insulin, and specifically an insulin analog with an improved in vitro effect compared with that of native insulin.
As used herein, the term "insulin analog" refers to a modified analog of native insulin prepared via modification of a part of the amino acid(s) of native insulin in the form of insertion, deletion, or substitution, and in particular, it includes various insulin analogs of native insulin with an improved in vitro effect compared with that of native insulin.
Native insulin is a hormone secreted by the pancreas and generally plays a role in promoting intracellular glucose absorption and inhibiting fat breakdown, thereby controlling in vivo blood glucose levels. Insulin is generated from the processing of its precursor, proinsulin, which does not have the function of controlling blood glucose levels. Insulin is composed of two polypeptide chains, i.e., the A-chain and the B-chain, which include 21 and 30 amino acids, respectively, and are interlinked by a disulfide bridge. Each of the A-chain and the B-chain may include the amino acid sequences represented by SEQ ID NO: 1 and SEQ ID NO: 2 shown below.
A-chain:
Gly-Ile-Val-Glu-Gln-Cys-Cys-Thr-Ser-Ile-Cys-Ser-Leu-Tyr-Gln-Leu-Glu-Asn-Tyr-Cys-Asn (SEQ ID NO: 1)
B-chain:
Phe-Val-Asn-Gln-His-Leu-Cys-Gly-Ser-His-Leu-Val-Glu-Ala-Leu-Tyr-Leu-Val-Cys-Gly-Glu-Arg-Gly-Phe-Phe-Tyr-Thr-Pro-Lys-Thr (SEQ ID NO: 2)
The insulin according to the present invention refers to insulin analogs prepared by genetic recombination technology, but the insulin is not limited thereto and includes all insulin with an improved in vitro effect compared with that of native insulin. Preferably, the insulin of the present invention includes inverted insulin, insulin variants, insulin fragments, etc. The insulin can be prepared not only by a recombinant method but also by a solid phase synthesis, and the preparation method is not limited thereto.
These insulin analogs, being peptides having an in vivo blood glucose level-controlling capability equivalent or corresponding to that of native insulin, include all of insulin agonists, insulin derivatives, insulin fragments, insulin variants, etc.
As used herein, the term "insulin agonist" refers to a material which can bind to an in vivo receptor of insulin regardless of the structure of insulin and thereby exhibit a biological activity equivalent to that of insulin.
As used herein, the term "insulin derivative" may refer to a peptide which has a homology to each of the amino acid sequences of the A-chain and the B-chain of native insulin and is in the form having an in vivo blood glucose level-controlling capability, where a part of the groups in an amino acid residue is modified by chemical substitution (e.g., alpha-methylation, alpha-hydroxylation), deletion (e.g., deamination), or modification (e.g., N-methylation).
Additionally, as used herein, the term "insulin derivative" may refer to a peptide mimic and a low or high molecular weight compound which can control in vivo blood glucose levels by binding to an insulin receptor, although there is no sequence homology to the amino acid sequence of native insulin.
As used herein, the term "insulin fragment" refers to a form of insulin in which at least one amino acid is inserted or deleted, and the amino acid inserted may be one that is not present in nature (e.g., D-type amino acid), and the insulin fragment has an in vivo blood glucose level-controlling capability.
As used herein, the term "insulin variant" refers to a peptide which has a difference in at least one amino acid sequence from that of insulin, and the peptide also has the in vivo blood glucose level-controlling capability.
The methods used in preparing insulin agonists, derivatives, fragments, and variants may be used independently or in combination. For example, those peptides having the in vivo blood glucose level-controlling capability, which have a difference in at least one amino acid sequence, and in which the amino acid residue in the N-terminus is deaminated, may be included in the scope of the present invention.
The insulin analog according to the present invention exclusively includes any peptide with an improved in vitro effect compared with that of native insulin by introducing substitution, insertion, or deletion of amino acid(s), or a post-translational modification (e.g., methylation, acylation, ubiquitination, and intermolecular covalent bond) in the amino acid sequences (SEQ ID NOS: 1 and 2) of the A-chain and the B-chain of native insulin. For the substitution or insertion of the amino acid(s), not only the 20 amino acids conventionally observed in human proteins but also atypical or unnatural amino acids may be used. The atypical amino acids may be commercially obtained from Sigma-Aldrich, ChemPep, Genzymepharmaceuticals, etc. The peptides containing these amino acids and typical peptide sequences may be synthesized by or purchased from commercial peptide synthesis companies, such as American Peptide Company, Bachem (USA), and Anygen (Korea).
Specifically, the insulin analogs according to the present invention may be those which include a modification or deletion in particular amino acid residue(s) of the A-chain and the B-chain of native insulin, and preferably, may be those in which particular amino acid residue(s) of the A-chain of native insulin is(are) modified and particular amino acid residue(s) of the B-chain of native insulin is(are) modified and/or deleted.
Preferably, the insulin analogs of the present invention may be an analog in which the 14th amino acid residue, tyrosine, in the amino acid sequence of the A-chain represented by SEQ ID NO: 1 is substituted with glutamic acid, asparagine, or alanine, or an analog in which the 16th amino acid residue, tyrosine, is substituted with glutamic acid and/or the 25th amino acid residue, phenylalanine, in the amino acid sequence of the B-chain represented by SEQ ID NO: 2 is deleted; or may include all of these.
More preferably, the insulin analogs of the present invention may be those which include the A-chain of SEQ ID NO: 3 represented by the following General Formula 1 and the B-chain of SEQ ID NO: 4 represented by the following General Formula 2.
General Formula 1
Xaa1-Ile-Val-Glu-Xaa2-Cys-Cys-Thr-Ser-Ile-Cys-Xaa3-Leu-Xaa4-Gln-Xaa5-Glu-Asn-Xaa6-Cys-Xaa7 (SEQ ID NO: 3)
In the above General Formula 1, Xaa1 is alanine, glycine, glutamine, histidine, glutamic acid, or asparagine;
Xaa2 is alanine, glutamic acid, glutamine, histidine, or asparagine;
Xaa3 is alanine, serine, glutamine, glutamic acid, histidine, or asparagine;
Xaa4 is alanine, tyrosine, glutamic acid, histidine, lysine, aspartic acid, or asparagine;
Xaa5 is alanine, leucine, tyrosine, histidine, glutamic acid, or asparagine;
Xaa6 is alanine, tyrosine, serine, glutamic acid, histidine, or asparagine; and
Xaa7 is asparagine, glycine, histidine, or alanine.
General Formula 2
Phe-Val-Asn-Gln-His-Leu-Cys-Gly-Ser-His-Leu-Val-Glu-Ala-Leu-Xaa8-Leu-Val-Cys-Gly-Glu-Arg-Gly-Phe-Xaa9-Tyr-Xaa10-Xaa11-Lys-Thr (SEQ ID NO: 4)
In the above General Formula 2,
Xaa8 is tyrosine, glutamic acid, or aspartic acid, or is absent;
Xaa9 is phenylalanine, or is absent;
Xaa10 is threonine, or is absent; and
Xaa11 is proline, glutamic acid, or aspartic acid, or is absent;
(wherein the peptides including the A-chain of SEQ ID NO: 1 and the B-chain of SEQ ID NO: 2 may be excluded).
In a more specific exemplary embodiment, the insulin ananlog may be an insulin analog, wherein, in General Formula 1,
Xaa1 is glycine,
Xaa2 is glutamine,
Xaa3 is serine,
Xaa4 is alanine, glutamic acid, or asparagine,
Xaa5 is leucine,
Xaa6 is tyrosine, and
Xaa7 is asparagine; and
in General Formula 2,
Xaa8 is tyrosine or glutamic acid,
Xaa9 is phenylalanine, or is absent,
Xaa10 is threonine, and
Xaa11 is proline, glutamic acid, or aspartic acid, or is absent,
but is not limited thereto.
In another specific exemplary embodiment, the insulin ananlog may be an insulin analog, wherein, in General Formula 1,
Xaa1 is glycine,
Xaa2 is glutamine,
Xaa3 is serine,
Xaa4 is glutamic acid or asparagine,
Xaa5 is leucine,
Xaa6 is tyrosine, and
Xaa7 is asparagine; and
in General Formula 2,
Xaa8 is tyrosine,
Xaa9 is phenylalanine, or is absent,
Xaa10 is threonine, and
Xaa11 is proline, glutamic acid, or aspartic acid, or is absent,
but is not limited thereto.
In still another specific exemplary embodiment, the insulin ananlog may include an A-chain of General Formula 1 and a B-chain, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
The insulin ananlog may include an A-chain of General Formula 1 and a B-chain, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is absent, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
The insulin ananlog may be characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
The insulin ananlog may be characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is asparagine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
In still another specific exemplary embodiment, the insulin ananlog may be characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or is absent, but is not limited thereto.
In still another specific exemplary embodiment, the insulin ananlog may be characterized in that, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is alanine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and in the B-chain of SEQ ID NO: 4, Xaa8 is glutamic acid, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline, glutamic acid, or aspartic acid, or absent, but is not limited thereto.
In an exemplary embodiment, the insulin analogs of the present invention may include the following analogs:
i) insulin analog 1: A peptide in which the 14th amino acid residue in the amino acid sequence of the A-chain represented by SEQ ID NO: 3 is glutamic acid and the 25th amino acid residue in the amino acid sequence of the B-chain represented by SEQ ID NO: 4 is phenylalanine, having an amino acid sequence represented by SEQ ID NO: 16 which is encoded by a nucleic acid containing a nucleotide sequence represented by SEQ ID NO: 15.
ii) insulin analog 2: A peptide in which the 14th amino acid residue in the amino acid sequence of the A-chain represented by SEQ ID NO: 3 is asparagine and the 25th amino acid residue in the amino acid sequence of the B-chain represented by SEQ ID NO: 4 is phenylalanine, having an amino acid sequence represented by SEQ ID NO: 18 which is encoded by a nucleic acid containing a nucleotide sequence represented by SEQ ID NO: 17.
iii) insulin analog 3: A peptide in which the 14th amino acid residue in the amino acid sequence of the A-chain represented by SEQ ID NO: 3 is glutamic acid and the 25th amino acid residue in the amino acid sequence of the B-chain represented by SEQ ID NO: 4 is deleted, having an amino acid sequence represented by SEQ ID NO: 20 which is encoded by a nucleic acid containing a nucleotide sequence represented by SEQ ID NO: 19.
iv) insulin analog 4: A peptide in which the 14th amino acid residue in the amino acid sequence of the A-chain represented by SEQ ID NO: 3 is alanine and the 16th amino acid residue in the amino acid sequence of the B-chain represented by SEQ ID NO: 4 is glutamic acid, and the 25th amino acid residue is absent, having an amino acid sequence represented by SEQ ID NO: 22 which is encoded by a nucleic acid containing a nucleotide sequence represented by SEQ ID NO: 21.
As used herein, the term "in vitro effect" refers to glucose uptake by an insulin analog, and it is indicated by the measurement result of EC50 on glucose uptake regarding mouse-derived 3T3-L1 cells differentiated into adipocytes.
In an exemplary embodiment, when the in vitro effect of insulin analogs 1 to 3 was measured, the insulin analog 1 showed a 238.4% increase of glucose uptake, the insulin analog 2 showed a 241.7% increase, and the insulin analog 3 showed a 705% increase compared with that of native insulin, respectively, thereby confirming that the insulin analogs according to the present invention exhibit a remarkable in vitro effect of a 2- to 7-fold increase compared with that of native insulin (Table 1).
In another aspect, the present invention provides nucleic acids encoding the above insulin analogs.
As used herein, the term "nucleic acid" refers to a deoxyribonucleotide (DNA) or ribonucleotide (RNA) including genomic DNA, cDNA, and RNA being transcribed therefrom, and a nucleotide as the basic constituting unit not only includes natural nucleotides but also includes analogues having modifications in a sugar or base (Scheit, Nucleotide Analogs, John Wiley, New York, 1980; Uhlman and Peyman, Chemical Reviews, 90: 543-584, 1990). The nucleic acid of the present invention may be isolated or prepared using standard technology in molecular biology. For example, the nucleic acid of the present invention may be prepared by PCR amplification using appropriate primer sequences based on the gene sequence of native insulin (NM_000207.2, NCBI), and may be prepared by standard synthesis technology using an automated DNA synthesizer.
Preferably, the nucleic acid of the present invention includes the nucleotide sequences represented by SEQ ID NOS: 15, 17, 19, and 21. The nucleic acid of the present invention not only includes the nucleotide sequences represented by SEQ ID NOS: 15, 17, 19, and 21, but also includes all the sequences which have a sequence homology of at least 70% to the above sequences, preferably at least 80%, more preferably at least 90%, even more preferably at least 95%, and most preferably at least 98%, and the peptide encoded by the above nucleic acid can bind to in vivo receptors of insulin, thereby exhibiting a biological activity substantially the same as that of insulin.
As used herein, the term "homology" refers to a degree of similarity with a given amino acid sequence of a native wild-type protein or a polynucleotide sequence encoding the same, and includes those sequences which have the identity of the above-described percentages or higher to the amino acid sequences or polynucleotide sequences of the present invention. The homology may be determined by comparing the two given sequences by the naked eye or may be determined using a bioinformatic algorithm which enables the analysis of a homology by arranging the subject sequences for comparison. The homology between the two given amino acid sequences may be indicated as a percentage. The useful automated algorithm is available for use in GAP, BESTFIT, FASTA, and TFASTA computer software modules of Wisconsin Genetics Software Package (Genetics Computer Group, Madison, WI, USA). The arrangement algorithm automated in the above modules includes sequence arrangement algorithms by Needleman & Wunsch, Pearson & Lipman, and Smith & Waterman. Other useful algorithms on sequence arrangement and homology determination are automated in software including FASTP, BLAST, BLAST2, PSIBLAST, and CLUSTAL W.
In another aspect, the present invention provides a recombinant vector including a nucleic acid encoding the insulin analog. The recombinant vector according to the present invention may be constructed as a vector for conventional cloning or expression, and may be constructed as a vector to use a prokaryotic cell or a eukaryotic cell as a host cell.
As used herein, the term "vector" refers to a recombinant vector capable of expressing a target protein in an appropriate host cell, which is a gene construct including essential regulatory factors operably linked to enable the expression of a nucleic acid insert. The present invention can prepare a recombinant vector which includes a nucleic acid encoding an insulin analog, and the insulin analog of the present invention may be obtained via transformation or transfection of the recombinant vector into a host cell.
In the present invention, the nucleic acid encoding the insulin analog is operably linked to a promoter. As used herein, the term "operably linked" refers to a functional connection between a regulatory sequence for nucleic acid expression (e.g., a promoter, a signal sequence, a ribosome-binding site, a transcription termination sequence, etc.) and a different nucleotide sequence, and the regulatory sequence can regulate the transcription and/or translation of the different nucleotide sequence by the same.
As used herein, the term "promoter" refers to an untranslated nucleic acid sequence located upstream of a coding region, which includes a polymerase-binding site and has the activity of initiating transcription of a gene located downstream of a promoter into mRNA, i.e., a DNA domain to which polymerase binds and initiates the transcription of a gene, and it is located at the 5' domain of mRNA transcription initiation.
For example, when the vector of the present invention is a recombinant vector and uses a prokaryotic cell as a host cell, in general, a strong promoter (e.g., tac promoter, lac promoter, lacUV5 promoter, lpp promoter, pL λ promoter, pRλ promoter, rac5 promoter, amp promoter, recA promoter, SP6 promoter, trp promoter, T7 promoter, etc.) capable of executing transcription, a ribosome-binding site for the initiation of translation, and transcription/translation termination sequences should be included.
Additionally, the vector to be used in the present invention may be prepared by manipulating the plasmids (e.g., pSC101 , pGV1106 , pACYC177 , ColE1, pKT230 , pME290 , pBR322 , pUC8 /9, pUC6 , pBD9 , pHC79 , pIJ61 , pLAFR1 , pHV14, pGEX series, pET series, pPICZα series, pUC19, etc.), phages (e.g., λgt4·λB, λ- Charon , λ△z1, M13, etc.), or viruses (e.g., SV40, etc.) which are commonly used in the art.
Meanwhile, when the vector of the present invention is a recombinant vector and uses a eukaryotic cell as a host cell, promoters derived from the genomes of mammalian cells (e.g., metallothionein promoter) or promoters derived from the mammalian viruses (e.g., adenovirus late promoter, 7.5K promoter of papillomavirus, SV40 promoter, cytomegalovirus promoter, and tk promoter of HSV) may be used, and in general, the vector includes a polyadenylated sequence (e.g., bovine growth hormone terminator and a polyadenylated sequence derived from SV40) as a transcription termination sequence.
Additionally, the recombinant vector of the present invention includes an antibiotic-resistance gene commonly used in the art as a selective marker, and may include, for example, genes having resistance to ampicillin, gentamycin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin, neomycin, and tetracycline.
The recombinant vector of the present invention may additionally include a different sequence to make it easy to purify target proteins being collected, i.e., a single-chain insulin analog, proinsulin, or an analog thereof. The sequence to be additionally included may be a tag sequence for protein purification, e.g., glutathione S-transferase (Pharmacia, USA), a maltose-binding protein (NEB, USA), FLAG (IBI, USA), 6-histidine, etc., but the kinds of the sequence necessary for the purification of target proteins are not limited thereto.
Fusion proteins expressed by the recombinant vector including the above tag sequence may be purified by affinity chromatography. For example, when glutathione S-transferase is fused, glutathione, which is the substrate of the enzyme, may be used, and when 6-histidine tag is used, a desired target protein may be easily collected by a Ni-NTA column.
In still another aspect, the present invention provides a transformant transformed by a recombinant vector including the nucleic acid encoding the insulin analog.
As used herein, the term "transformation" refers to a process of introducing DNA into a host cell and making the DNA replicable therein as a chromosomal factor or by completion of chromosomal integration, which is a phenomenon of artificially causing a genetic change by introducing exogenous DNA into a cell.
The method of transformation used in the present invention may be any transformation method, and it may be easily performed according to the conventional method used in the art. Examples of the commonly used transformation method may include a CaCl2 precipitation method, the Hanahan method with improved efficiency using dimethyl sulfoxide (DMSO) as a reducing agent in the CaCl2 precipitation method, electroporation, a CaPO4 precipitation method, a protoplast fusion method, a stirring method using silicon carbide fiber, an agrobacteria-mediated transformation, a transformation using PEG, dextran sulfate-, lipofectamine-, and dry/suppression-mediated transformations, etc.
The method for transforming the recombinant vector including a nucleic acid encoding an insulin analog according to the present invention may not be limited to these methods, but any method for transformation or transfection commonly used in the art may be used without limitation.
The transformant of the present invention may be obtained by introducing a recombinant vector including the target nucleic acid which encodes an insulin analog into a host cell.An appropriate host to be used in the present invention may not be particularly limited as long as it can express the nucleic acid of the present invention. Examples of the appropriate host may include a bacteria belonging to the genus Escherichia such as E. coli, a bacteria belonging to the genus Bacillus such as Bacillus subtilis, a bacteria belonging to the genus Pseudomonas such as Pseudomonas putida, yeasts such as Pichia pastoris, Saccharomyces cerevisiae, and Schizosaccharomyces pombe, an insect cell such as Spodoptera frugiperda (SF9), and animal cells such as CHO, COS, and BSC. Preferably, E. coli is used as a host cell.
In an exemplary embodiment, the respective nucleotide sequence encoding the insulin analogs 1 to 3 according to the present invention was amplified via PCR, and the amplified gene fragments were cloned into pET22b vector (Novagen). For the expression of the insulin analogs in the form of an inclusion body in a cell, the pET22b vector was treated with restriction enzymes, NdeI and BamHI, to remove a signal sequence therein, the PCR-amplified products of the insulin analogs were treated with the same restriction enzymes, NdeI and BamHI, and the respective isolated DNA was inserted into the pET22b cloning vector using T4 DNA ligase. The thus-obtained expression vectors were named as pET22b-insulin analogs 1 to 4, respectively.
The expression vector pET22b-insulin analogs 1 to 4 respectively encode amino acid sequences represented by SEQ ID NOS: 16, 18, 20, and 22, under the control of T7 promoter, and each of the insulin analogs was expressed in the form of an inclusion body in a host cell, respectively.
The recombinant vector pET22b-insulin analogs 1 to 4 including nucleic acids encoding each of the insulin analogs of SEQ ID NOS: 16, 18, 20, and 22 were transformed into E. coli, respectively, and thereby transformants expressing them in the form of an inclusion body were obtained.
In still another aspect, the present invention provides a method for preparing an insulin analog using the transformants.
Preferably, the present invention provides a method for preparing an insulin analog, including:
a) preparing a recombinant expression vector including a nucleic acid encoding the insulin analog;
b) transforming the recombinant expression vector into a host cell and obtaining a transformant therefrom;
c) culturing the transformant and expressing the insulin analog; and
d) isolating and purifying the expressed insulin analog peptide.
The medium used in culturing the transformants in the present invention should meet the requirements for host cell cultivation in an appropriate manner. The carbon sources to be contained in the medium for the growth of a host cell may be appropriately selected by the decision of a skilled person in the art according to the transformants prepared thereof, and appropriate cultivation conditions may be selected to control the period and amount of cultivation.
Examples of the sugar source to be used may include sugars and carbohydrates such as glucose, saccharose, lactose, fructose, maltose, starch, and cellulose; oils and fats such as soybean oil, sunflower oil, castor oil, and coconut oil; fatty acids such as palmitic acid, stearic acid, and linoleic acid; alcohols such as glycerol and ethanol; and organic acids such as acetic acid. These materials may be used alone or in combination.
Examples of the nitrogen source to be used may include peptone, yeast extract, meat gravy, malt extract, corn steep liquor, soybean flour, and urea, or inorganic compounds such as ammonium sulfate, ammonium chloride, ammonium phosphate, ammonium carbonate, and ammonium nitrate. The nitrogen source may also be used alone or in combination.
Examples of the phosphorous source to be used may include potassium dihydrogen phosphate or dipotassium hydrogen phosphate or a corresponding sodium-containing salt. Additionally, the culture media may contain a metal salt such as magnesium sulfate or iron sulfate necessary for the growth of the transformant. Furthermore, essential growth materials such as amino acids and vitamins may be used. Furthermore, appropriate precursors for culture media may also be used. The above sources may be appropriately added to a culture during cultivation by a batch culture or continuous culture. The pH of the culture may be appropriately adjusted using a basic compound such as sodium hydroxide, potassium hydroxide, and ammonia, or an acid compound such as phosphoric acid or sulfuric acid. Additionally, an antifoaming agent such as fatty acid polyglycol ester may be added to prevent foam generation. Additionally, in order to maintain the aerobic state of the culture, oxygen or an oxygen-containing gas (e.g., air) may be injected into the culture. The transformant of the present invention may be cultured at 20°C to 45°C, and preferably, 25°C to 40°C. Additionally, the cultivation is continued until the maximum amount of production of the desired insulin analogs is obtained, and in this regard, the cultivation may normally be continued for 10 hours to 160 hours.
As described above, the transformant of the present invention can produce insulin analogs when appropriate culture conditions are provided according to host cells, and the peptide-N-glycosidase produced thereof according to the vector constitution and characteristics of a host cell may be secreted within the cytoplasm or into the periplasmic space of the host cell or extracellularly.
The proteins expressed within or outside of the host cell may be purified by a conventional method.
Examples of the purification method may include salting-out (e.g., ammonium sulfate precipitation, ammonium phosphate precipitation, etc.), solvent precipitation (e.g., protein fraction precipitation using acetone or ethanol, etc.), dialysis, gel filtration, ion exchange, or chromatography such as reversed column chromatography, ultrafiltration, etc., and these methods may be used alone or in combination.
The transformant of the present invention is characterized in that the insulin analogs 1 to 3 are expressed from the recombinant vector pET22b-insulin analogs 1 to 3 in the form of an inclusion body under the control of T7 promoter. Accordingly, it is preferable that the insulin analogs 1 to 3, which were expressed in the form of an inclusion body, are converted into a soluble form and then isolated and purified.
In an exemplary embodiment, the present invention may further include the following steps for isolating and purifying the insulin analogs expressed in the form of an inclusion body from the transformant:
d-1) obtaining the transformant cells from the culture and pulverizing the same;
d-2) recovering the expressed insulin analog peptide from the pulverized cell lysate followed by refolding the same;
d-3) purifying the refolded insulin analog peptide by cation exchange chromatography;
d-4) treating the purified insulin analog peptide with trypsin and carboxypeptidase B; and
d-5) sequentially purifying the treated insulin analog peptide by cation exchange chromatography and anion exchange chromatography.
In still another aspect, the present invention provides a pharmaceutical composition for treating diabetes containing the above-mentioned insulin analogs.
The pharmaceutical composition containing the insulin analogs according to the present invention may contain a pharmaceutically acceptable carrier. Examples of the pharmaceutically acceptable carrier for oral administration may include a binder, a glidant, a disintegrating agent, an excipient, a solubilizing agent, a dispersing agent, a stabilizing agent, a suspending agent, a coloring agent, a flavoring agent, etc.; for injection formulations, a buffering agent, a preserving agent, an analgesic, an isotonic agent, a stabilizing agent, etc. may be mixed for use; and for topical formulations, a base, an excipient, a lubricant, a preserving agent, etc. may be used. The formulation type of the pharmaceutical composition according to the present invention may be prepared variously by combination with the pharmaceutically acceptable carriers described above. For example, for oral administration, the pharmaceutical composition may be formulated into tablets, troches, capsules, elixirs, suspensions, syrups, wafers, etc. For injections, the pharmaceutical composition may be formulated into single-dose ampoules or multidose containers. Additionally, the pharmaceutical composition may also be formulated into solutions, suspensions, tablets, pills, capsules, and sustained-release formulations.
Meanwhile, examples of suitable carriers, excipients, and diluents may include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinylpyrrolidone, water, methyl hydroxybenzoate, propyl hydroxybenzoate, talc, magnesium stearate, mineral oil, etc. Additionally, the pharmaceutical composition of the present invention may further contain a filler, an anti-coagulant, a lubricant, a humectant, a flavoring agent, an emulsifier, a preservative, etc.
In still another aspect, the present invention provides a method for treating diabetes including administering a pharmaceutical composition containing the insulin analogs of the present invention to a subject in need thereof.
The insulin analogs according to the present invention exhibit a significantly improved in vitro effect compared with that of native insulin, and thus it is expected that the administration of a pharmaceutical composition containing the above insulin analogs can be effective for treating diabetes.
As used herein, the term "administration" refers to introduction of a particular material to a patient by an appropriate manner, and the conjugate of the present invention may be administered via any of the common routes as long as the drug can arrive at a target tissue. For example, intraperitoneal, intravenous, intramuscular, subcutaneous, intradermal, oral, topical, intranasal, intrapulmonary, and intrarectal administration may be performed, but the administration route is not limited thereto. However, since peptides are digested upon oral administration, active ingredients of a composition for oral administration should be coated or formulated for protection against degradation in the stomach. Preferably, the present composition may be administered in an injectable form. In addition, the pharmaceutical composition may be administered using a certain apparatus capable of transporting the active ingredients into a target cell.
Additionally, the pharmaceutical composition of the present invention may be determined by the types of the drug as an active component as well as by several related factors including the types of diseases to be treated, administration routes, age, sex, and weight of a patient, and severity of the illness. Since the pharmaceutical composition of the present invention has excellent in vivo duration and titer, it can considerably reduce the administration frequency and dose of pharmaceutical drugs of the present invention.
Hereinafter, the present invention will be described in more detail with reference to the following Examples. However, these Examples are for illustrative purposes only, and the invention is not intended to be limited by these Examples.
Example 1: Construction of an expression vector for insulin analogs
In order to construct insulin analogs in which amino acid(s) of the A-chain and/or the B-chain of native insulin were modified, primer pairs consisting of a forward primer and a reverse primer for amplifying the insulin analogs introduced with the corresponding modification were synthesized, and PCR was then performed using proinsulin cDNA as a template. In particular, the template used was that in which proinsulin cDNA (SC128255, OriGene) (see sequences: BC005255.1 and AAH05255) was cloned into pET22b vector (Novagen), and for smooth recombinant expression of insulin, the nucleotide sequence of SEQ ID NO: 23 (ATG GCA ACA ACA TCA ACA GCA ACT ACG CGT), which encodes the amino acid sequence of Met Ala Thr Thr Ser Thr Ala Thr Thr Arg (SEQ ID NO: 24), was inserted into the cloned proinsulin cDNA as a N-terminal fusion partner.
Specifically, in the present invention, the following insulin analogs including the amino acid modifications shown in Table 1 were synthesized.
Amino Acid Modification
Insulin Analog 1 A14 Tyr → Glu
Insulin Analog 2 A14 Tyr → Asn
Insulin Analog 3 A14 Tyr → Glu + B25 deletion
Insulin Analog 4 A14 Tyr → Ala + B16 Tyr → Glu, B25 deletion
In Table 1 above, the insulin analog 1 is an analog which includes a substitution of the 14th amino acid in the amino acid sequence of the A-chain of native insulin represented by SEQ ID NO: 1, i.e., tyrosine, with glutamic acid;
the insulin analog 2 is an analog which includes a substitution of the 14th amino acid in the amino acid sequence of the A-chain of native insulin represented by SEQ ID NO: 1, i.e., tyrosine, with asparagine;
the insulin analog 3 is an analog which includes a substitution of the 14th amino acid in the amino acid sequence of the A-chain of native insulin represented by SEQ ID NO: 1, i.e., tyrosine, with glutamic acid, and a deletion of the 25th amino acid in the amino acid sequence of the B-chain of native insulin represented by SEQ ID NO: 2, i.e., phenylalanine, and
the insulin analog 4 is an analog which includes a substitution of the 14th amino acid in the amino acid sequence of the A-chain of native insulin represented by SEQ ID NO: 1, i.e., tyrosine, with alanine, and a substitution of the 16th amino acid in the amino acid sequence of the B-chain of native insulin represented by SEQ ID NO: 2, i.e., tyrosine, with glutamic acid and a deletion of the 25th amino acid in the amino acid sequence of the B-chain of native insulin represented by SEQ ID NO: 2, i.e., phenylalanine.
The respective primer pairs of forward primers and reverse primers designed for the amplification of the insulin analogs 1 to 3 are shown in Table 2 below.
Sequence SEQ ID NO
Insulin Analog
1 5'-ccagcatctgctccctcgaacagctggagaactactg-3' 5
5'-cagtagttctccagctgttcgagggagcagatgctgg-3' 6
Insulin Analog 2 5'-cagcatctgctccctcaaccagctggagaactac-3' 7
5'-gtagttctccagctggttgagggagcagatgctg-3' 8
Insulin Analog 3 5'-ccagcatctgctccctcgaacagctggagaactactg-3' 5
5'-cagtagttctccagctgttcgagggagcagatgctgg-3' 6
5'-gcggggaacgaggcttctacacacccaagacccg-3' 9
5'-cgggtcttgggtgtgtagaagcctcgttccccgc-3' 10
Insulin Analog 4 5'-cagcatctgctccctcgcccagctggagaactac-3' 11
5'-gtagttctccagctgggcgagggagcagatgctg-3' 12
5'-ctggtggaagctctcgagctagtgtgcggggaac-3' 13
5'-gttccccgcacactagctcgagagcttccaccag-3' 14
5'-gcggggaacgaggcttctacacacccaagacccg-3' 9
5'-cgggtcttgggtgtgtagaagcctcgttccccgc-3' 10
In Table 2 above, the primer pair consisting of SEQ ID NOS: 5 and 6 was designed for the substitution of the 14th amino acid in the amino acid sequence of the A-chain of native insulin, i.e., tyrosine, with glutamic acid; the primer pair consisting of SEQ ID NOS: 7 and 8 was designed for the substitution of the 14th amino acid in the amino acid sequence of the A-chain of native insulin, i.e., tyrosine, with asparagine; the primer pair consisting of SEQ ID NOS: 9 and 10 was designed for the deletion of the 25th amino acid in the amino acid sequence of the B-chain of native insulin, i.e., phenylalanine; the primer pair consisting of SEQ ID NOS: 11 and 12 was designed for the substitution of the 14th amino acid in the amino acid sequence of the A-chain of native insulin i.e., tyrosine, with alanine; and the primer pair consisting of SEQ ID NOS: 13 and 14 was designed for the substitution of the 16th amino acid in the amino acid sequence of the B-chain of native insulin, i.e., tyrosine, with glutamic acid
In order to perform PCR for the amplification of insulin analogs which include the corresponding modifications, a reaction solution was prepared by mixing 150 ng of template DNA, 1 mL each of 100 pM primers, 5 mL of 2.5 mM dNTP, 10 units of pfx polymerase (Invitrogen, USA), and a 10X buffer solution. The reaction solution was subjected to initial denaturation at 95°C for 30 seconds, followed by 18 repeated cycles of annealing at 95°C for 30 seconds, 55°C for 30 seconds, and 68°C for 6 minutes, and it was finally left at 68°C for 5 minutes. The thus-obtained PCR-amplified products were extracted using a gel extraction kit (Qiagen, Germany) and treated with restriction enzymes, NdeI and BamHI, to prepare insertion fragments. The pET22b vector (Novagen, USA) was then cleaved with the same restriction enzymes and fragments were extracted using the same gel extraction kit. The above insertion fragments were ligated into the thus-prepared vector using T4 ligase to prepare expression vector pET22b-insulin analogs 1 to 4. The expression vectors include nucleic acids encoding the amino acid sequences of the insulin analogs 1 to 4 under the control of T7 promoter, and the vectors can express the insulin analog proteins in the form of an inclusion body in a host cell.
The thus-obtained expression vector pET22b-insulin analog 1 according to the present invention includes nucleic acid having a nucleotide sequence represented by SEQ ID NO: 15, which encodes the insulin analog having an amino acid sequence represented by SEQ ID NO: 16; the thus-obtained expression vector pET22b-insulin analog 2 according to the present invention includes a nucleic acid having a nucleotide sequence represented by SEQ ID NO: 17 which encodes the insulin analog having an amino acid sequence represented by SEQ ID NO: 18; the thus-obtained expression vector pET22b-insulin analog 3 according to the present invention includes a nucleic acid having a nucleotide sequence represented by SEQ ID NO: 19, which encodes the insulin analog having an amino acid sequence represented by SEQ ID NO: 20; and the thus-obtained expression vector pET22b-insulin analog 4 according to the present invention includes a nucleic acid having a nucleotide sequence represented by SEQ ID NO: 21, which encodes the insulin analog having an amino acid sequence represented by SEQ ID NO: 22
The DNA sequences and protein sequences of each of the insulin analogs 1 to 4 are shown in Table 3 below.
Sequence SEQ ID NO
Insulin Analog 1 DNA TTC GTT AAC CAA CAC TTG TGT GGC TCA CAC CTG GTG GAA GCT CTC TAC CTA GTG TGC GGG GAA CGA GGC TTC TTC TAC ACA CCC AAG ACC CGC CGG GAG GCA GAG GAC CTG CAG GTG GGG CAG GTG GAG CTG GGC GGG GGC CCT GGT GCA GGC AGC CTG CAG CCC TTG GCC CTG GAG GGG TCC CTG CAG AAG CGT GGC ATT GTG GAA CAA TGC TGT ACC AGC ATC TGC TCC CTC GAA CAG CTG GAG AAC TAC TGC AAC TGA 15
Protein Phe Val Asn Gln His Leu Cys Gly Ser His Leu Val Glu Ala Leu Tyr Leu Val Cys Gly Glu Arg Gly Phe Phe Tyr Thr Pro Lys Thr Arg Arg Glu Ala Glu Asp Leu Gln Val Gly Gln Val Glu Leu Gly Gly Gly Pro Gly Ala Gly Ser Leu Gln Pro Leu Ala Leu Glu Gly Ser Leu Gln Lys Arg Gly Ile Val Glu Gln Cys Cys Thr Ser Ile Cys Ser Leu Glu Gln Leu Glu Asn Tyr Cys Asn 16
Insulin Analog 2 DNA TTC GTT AAC CAA CAC TTG TGT GGC TCA CAC CTG GTG GAA GCT CTC TAC CTA GTG TGC GGG GAA CGA GGC TTC TTC TAC ACA CCC AAG ACC CGC CGG GAG GCA GAG GAC CTG CAG GTG GGG CAG GTG GAG CTG GGC GGG GGC CCT GGT GCA GGC AGC CTG CAG CCC TTG GCC CTG GAG GGG TCC CTG CAG AAG CGT GGC ATT GTG GAA CAA TGC TGT ACC AGC ATC TGC TCC CTC AAC CAG CTG GAG AAC TAC TGC AAC TGA 17
Protein Phe Val Asn Gln His Leu Cys Gly Ser His Leu Val Glu Ala Leu Tyr Leu Val Cys Gly Glu Arg Gly Phe Phe Tyr Thr Pro Lys Thr Arg Arg Glu Ala Glu Asp Leu Gln Val Gly Gln Val Glu Leu Gly Gly Gly Pro Gly Ala Gly Ser Leu Gln Pro Leu Ala Leu Glu Gly Ser Leu Gln Lys Arg Gly Ile Val Glu Gln Cys Cys Thr Ser Ile Cys Ser Leu Asn Gln Leu Glu Asn Tyr Cys Asn 18
Insulin Analog 3 DNA TTC GTT AAC CAA CAC TTG TGT GGC TCA CAC CTG GTG GAA GCT CTC TAC CTA GTG TGC GGG GAA CGA GGC TTC TAC ACA CCC AAG ACC CGC CGG GAG GCA GAG GAC CTG CAG GTG GGG CAG GTG GAG CTG GGC GGG GGC CCT GGT GCA GGC AGC CTG CAG CCC TTG GCC CTG GAG GGG TCC CTG CAG AAG CGT GGC ATT GTG GAA CAA TGC TGT ACC AGC ATC TGC TCC CTC GAA CAG CTG GAG AAC TAC TGC AAC TGA 19
Protein Phe Val Asn Gln His Leu Cys Gly Ser His Leu Val Glu Ala Leu Tyr Leu Val Cys Gly Glu Arg Gly Phe Tyr Thr Pro Lys Thr Arg Arg Glu Ala Glu Asp Leu Gln Val Gly Gln Val Glu Leu Gly Gly Gly Pro Gly Ala Gly Ser Leu Gln Pro Leu Ala Leu Glu Gly Ser Leu Gln Lys Arg Gly Ile Val Glu Gln Cys Cys Thr Ser Ile Cys Ser Leu Glu Gln Leu Glu Asn Tyr Cys Asn 20
Insulin Analog 4 DNA TTC GTT AAC CAA CAC TTG TGT GGC TCA CAC CTG GTG GAA GCT CTC GAG CTA GTG TGC GGG GAA CGA GGC TTC TAC ACA CCC AAG ACC CGC CGG GAG GCA GAG GAC CTG CAG GTG GGG CAG GTG GAG CTG GGC GGG GGC CCT GGT GCA GGC AGC CTG CAG CCC TTG GCC CTG GAG GGG TCC CTG CAG AAG CGT GGC ATT GTG GAA CAA TGC TGT ACC AGC ATC TGC TCC CTC GCC CAG CTG GAG AAC TAC TGC AAC TGA 21
Protein Phe Val Asn Gln His Leu Cys Gly Ser His Leu Val Glu Ala Leu Glu Leu Val Cys Gly Glu Arg Gly Phe Tyr Thr Pro Lys Thr Arg Arg Glu Ala Glu Asp Leu Gln Val Gly Gln Val Glu Leu Gly Gly Gly Pro Gly Ala Gly Ser Leu Gln Pro Leu Ala Leu Glu Gly Ser Leu Gln Lys Arg Gly Ile Val Glu Gln Cys Cys Thr Ser Ile Cys Ser Leu Ala Gln Leu Glu Asn Tyr Cys Asn 22
Example 2: Expression of recombinant insulin analogs
The recombinant expression of insulin analogs according to the present invention under the control of T7 promoter was performed as follows. E . coli BL21-DE3 (E. coli B F-dcm ompT hsdS(rB-mB-) gal λDE3) (Novagen, USA) was transformed with each of the insulin analog expression vectors prepared in Example 1. Transformation was performed using a method recommended by Novagen, the manufacturer of E. coli BL21- DE3. Each single colony transformed with the insulin analog expression vectors was collected, inoculated into a 2X Luria Broth (LB) medium containing 50 μg/mL ampicillin, and cultured at 37°C for 15 hours. The recombinant E. coli culture and the 2X LB medium containing 30% glycerol were mixed in a 1:1 (v/v) ratio, aliquoted 1 mL of the mixture into each cryo-tube, respectively, and stored at -140°C. The resultant was used as a cell stock for producing recombinant insulin analogs.
For the expression of recombinant insulin analogs, one vial of each cell stock was dissolved in 500 mL of 2X LB and incubated in a shaking water bath maintained at 37°C for 14 hours to 16 hours. The incubation was stopped when the OD value reached 5.0 or higher, and the culture was used as a seed culture. The seed culture was inoculated into 17 L of a fermentation medium using a 50 L fermenter (MSJ-U2, B.E. MARUBISHI, Japan) and the initial batch fermentation was started. The cultivation was performed at 37°C at a stirring rate of 500 rpm with 20 L/min (1 vvm) of air supply while maintaining the pH at 6.70 with 30% ammonia water. Regarding the progress of the fermentation, when the nutrients in the culture medium were limited, the fermentation was carried out in a fed-batch culture by adding a feeding solution. The growth of bacteria was monitored based on OD values, and when the OD value reached 100 or higher, IPTG at a final concentration of 500 μM was introduced therein. The cultivation was continued further for about 23 hours to 25 hours after the introduction. Upon termination of the cultivation, the recombinant bacteria was recovered by centrifugation and stored at -80°C until use.
Example 3: Isolation and purification of recombinant insulin analogs
For the isolation and purification of the recombinant insulin analogs expressed in Example 2 from the transformants, cells were disrupted as shown below followed by refolding in order to change the insulin analogs expressed in the form of a water-insoluble inclusion body to a water-soluble form.
<3-1> Recovery and refolding of recombinant insulin analogs
Specifically, each cell pellet was resuspended in a 1 L solubilizing buffer solution (50 mM Tris-HCl (pH 9.0), 1 mM EDTA (pH 8.0), 0.2 M NaCl, and 0.5% Triton X-100), and the cells were disrupted using a microfluidizer M-110EH (AC Technology Corp. Model M1475C) at a pressure of 15,000 psi. The disrupted cell lysates were centrifuged at 7,000 rpm and 4°C for 20 minutes and the supernatant was discarded. The resultant was resuspended in 3 L of a washing buffer (0.5% Triton X-100, 50 mM Tris (pH 8.0), 0.2 M NaCl, and 1 mM EDTA). Centrifugation was performed at 7,000 rpm and 4°C for 20 minutes, and the resulting pellet was resuspended in distilled water, followed by centrifugation in the same manner. Each of the resulting pellets was resuspended in 400 mL of a buffer solution (1 M glycine, 3.78 g cysteine-HCl, pH 10.6) and stirred at room temperature for 1 hour. In order to recover the resuspended recombinant insulin analogs, 400 mL of 8 M urea was added thereto and stirred at 40°C for 1 hour. For the refolding of the solubilized recombinant insulin analogs, the resultant was centrifuged at 7,000 rpm and 4°C for 20 minutes, and the supernatant was recovered. The supernatant was stirred at 4°C for 16 hours while 7.2 L of distilled water was added using a peristaltic pump at a flow rate of 1000 mL/hour.
<3-2> Purification of cation exchange chromatography
The samples in which the refolding was completed in Example <3-1> were respectively loaded into a cation exchange column (Source S, GE Healthcare), which was equilibrated with a 20 mM sodium citrate buffer solution (pH 2.0) containing 45% ethanol to be conjugated thereto. Insulin analog proteins were then eluted from the column with a linear concentration gradient from 0% to 100% in 10 column volumes using a 20 mM sodium citrate buffer solution (pH 2.0) which contained 0.5 M potassium chloride and 45% ethanol.
<3-3> Treatment with trypsin and carboxypeptidase B
Salts were removed from the samples eluted in Example <3-2> using a desalting column, followed by replacement of a buffer solution (10 mM Tris-HCl, pH 8.0). The samples were treated with trypsin, which corresponds to a molar ratio of 1000 relative to the protein amount of the sample, and carboxypeptidase B, which corresponds to a molar ratio of 2000 relative to the protein amount of the sample, and stirred at 16°C for 16 hours. The reaction was stopped by lowering the pH to 3.5 using 1 M sodium citrate (pH 2.0).
<3-4> Purification of cation exchange chromatography
The samples in which the reaction was completed in Example <3-3> were respectively reloaded into a cation exchange column (Source S, GE Healthcare) which was equilibrated with a 20 mM sodium citrate buffer solution (pH 2.0) containing 45% ethanol to be conjugated thereto. Insulin analog proteins were then eluted from the column with a linear concentration gradient from 0% to 100% in 10 column volumes using a 20 mM sodium citrate buffer solution (pH 2.0) which contained 0.5 M potassium chloride and 45% ethanol.
<3-5> Purification of anion exchange chromatography
Salts were removed from the samples eluted in Example <3-4> using a desalting column, followed by replacement of a buffer solution (10 mM Tris-HCl, pH 7.5). For the isolation of pure insulin analogs from the thus-obtained samples, the resultants were respectively loaded into an anion exchange column (Source Q, GE Healthcare) equilibrated with a 10 mM Tris buffer solution (pH 7.5) to be conjugated. Insulin analog proteins were then eluted from the column with a linear concentration gradient from 0% to 100% in 10 column volumes using a 10 mM Tris buffer solution (pH 7.5) which contained 0.5 M sodium chloride.
The purity of the purified insulin analogs was analyzed via protein electrophoresis (SDS-PAGE) and reversed phase and size exclusion chromatography, and the results are shown in FIG. 1 and FIG. 2, respectively. Additionally, the modifications in amino acids were confirmed by peptide mapping and the analysis of molecular weight of each peak, and the results are shown in FIG. 3.
As a result, it was confirmed that there was a modification in an amino acid sequence for each of the insulin analogs according to their desired purposes.
Example 4: Comparison of in vitro effect between native insulin and insulin analogs
In order to measure the in vitro effect of the insulin analogs isolated and purified in Example 3, an experiment on glucose absorption capability (glucose uptake or lipid synthesis capability) was performed using a mouse-derived 3T3-L1 cell line, which was differentiated into adipocytes. The 3T3-L1 cell line (ATCC, CL-173) was subcultured using Dulbecco's Modified Eagle's Medium (DMEM, Gibco, Cat. No. 12430) containing 10% bovine newborn calf serum (NBCS) two to three times per week. The 3T3-L1 cell line was suspended in a differentiation medium (DMEM containing 10% FBS), inoculated into a 48-well plate at a concentration of 5X104 cells/well, and cultured at 37°C for 48 hours. For the differentiation of the 3T3-L1 cell line into adipocytes, the differentiation medium was treated with 1 μg/mL of human insulin (Sigma, Cat. No. I9278), 0.5 mM IBMX (3-isobutyl-1-methylxanthine, Sigma, Cat. No. I5879), and 1 μM dexamethasone (Sigma, Cat. No. D4902), and the existing medium was removed and the mixture was aliquoted into each well in the amount of 250 μL/well. Forty-eight hours thereafter, the medium was replaced with a differentiation medium to which only 1 μg/mL of human insulin was added. The induction of differentiation of the 3T3-L1 cell line into adipocytes was then confirmed for a period of 7 to 9 days while replacing the medium with the differentiation medium containing 1 μg/mL of human insulin at 48 hour intervals.
For the experiment on glucose absorption capability, the cells which completed their differentiation into adipocytes were washed once with a serum-free DMEM medium, and then treated with 250 μL of the serum-free DMEM medium for 4 hours to induce serum depletion therein.
Human insulin and insulin analogs were respectively subjected to a 10-fold serial dilution from 5 μM to 0.005 nM using serum-free DMEM medium to be used as samples. The thus-prepared insulin samples were respectively added into cells in an amount of 250 μL, and cultured at 37°C for 24 hours in a 5% CO2 incubator. In order to measure the remaining glucose amount in the medium for which cultivation was completed, each culture sample was collected in an amount of 200 μL, diluted 5-fold using D-PBS, and subjected to the GOPOD analysis (GOPOD Assay Kit, Megazyme, Cat. No. K-GLUC). The concentration of the remaining glucose was calculated based on the absorbance of a glucose standard solution, the EC50 values on glucose uptake capability of the insulin analogs were respectively calculated, and the results are shown in Table 4 below.
Glucose Uptake Capability(relative to native insulin) (%)
Native Human Insulin 100
Insulin Analog 1 238.4
Insulin Analog 2 241.7
Insulin Analog 3 705
As shown in Table 4, the insulin analog 1 showed a 238.4% increase of glucose uptake capability, the insulin analog 2 showed a 241.7% increase, and the insulin analog 3 showed a 705% increase, compared with that of native insulin, respectively.
From the above results, it was confirmed that the insulin analogs according to the present invention exhibit a remarkable in vitro effect of a 2- to 7-fold increase compared with that of native insulin, and these results indicate that the insulin analogs can significantly increase their in vivo serum half-life and can thus be provided as stable insulin formulations, thus being effectively used as a therapeutic agent for treating diabetes.
Those of ordinary skill in the art will recognize that the present invention may be embodied in other specific forms without departing from its spirit or essential characteristics. The described embodiments are to be considered in all respects only as illustrative and not restrictive. The scope of the present invention is, therefore, indicated by the appended claims rather than by the foregoing description. All changes which come within the meaning and range of equivalency of the claims are to be embraced within the scope of the present invention.

Claims (21)

  1. An insulin analog peptide comprising an A-chain of SEQ ID NO: 3 indicated in the following General Formula 1 and a B-chain of SEQ ID NO: 4 indicated in the following General Formula 2:
    (General Formula 1)
    Xaa1-Ile-Val-Glu-Xaa2-Cys-Cys-Thr-Ser-Ile-Cys-Xaa3-Leu-Xaa4-Gln-Xaa5-Glu-Asn-Xaa6-Cys-Xaa7(SEQ ID NO: 3)
    wherein
    Xaa1 is alanine, glycine, glutamine, histidine, glutamic acid, or asparagine;
    Xaa2 is alanine, glutamic acid, glutamine, histidine, or asparagine;
    Xaa3 is alanine, serine, glutamine, glutamic acid, histidine, or asparagine;
    Xaa4 is alanine, tyrosine, glutamic acid, histidine, lysine, aspartic acid, or asparagine;
    Xaa5 is alanine, leucine, tyrosine, histidine, glutamic acid, or asparagine;
    Xaa6 is alanine, tyrosine, serine, glutamic acid, histidine, or asparagine; and
    Xaa7 is asparagine, glycine, histidine, or alanine; and
    (General Formula 2)
    Phe-Val-Asn-Gln-His-Leu-Cys-Gly-Ser-His-Leu-Val-Glu-Ala-Leu-Xaa8-Leu-Val-Cys-Gly-Glu-Arg-Gly-Phe-Xaa9-Tyr-Xaa10-Xaa11-Lys-Thr (SEQ ID NO: 4)
    wherein
    Xaa8 is tyrosine, glutamic acid, or aspartic acid, or is absent;
    Xaa9 is phenylalanine, or is absent;
    Xaa10 is threonine, or is absent; and
    Xaa11 is proline, glutamic acid, or aspartic acid, or is absent;
    (with the proviso that the peptide comprising the A-chain of SEQ ID NO: 1 and the B-chain of SEQ ID NO: 2 is excluded).
  2. The insulin analog peptide of claim 1, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, and Xaa10 is threonine.
  3. The insulin analog peptide of claim 1, wherein, in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, and Xaa10 is absent.
  4. The insulin analog peptide of claim 1,
    wherein, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and
    in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline.
  5. The insulin analog peptide of claim 4, wherein the peptide comprises an amino acid sequence represented by SEQ ID NO: 16.
  6. The insulin analog peptide of claim 1,
    wherein, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is asparagine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and
    in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is phenylalanine, Xaa10 is threonine, and Xaa11 is proline.
  7. The insulin analog peptide of claim 6, wherein the peptide comprises an amino acid sequence represented by SEQ ID NO: 18.
  8. The insulin analog peptide of claim 1,
    wherein, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is glutamic acid, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and
    in the B-chain of SEQ ID NO: 4, Xaa8 is tyrosine, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline.
  9. The insulin analog peptide of claim 8, wherein the peptide comprises an amino acid sequence represented by SEQ ID NO: 20.
  10. The insulin analog peptide of claim 1, wherein, in the A-chain of SEQ ID NO: 3, Xaa1 is glycine, Xaa2 is glutamine, Xaa3 is serine, Xaa4 is alanine, Xaa5 is leucine, Xaa6 is tyrosine, and Xaa7 is asparagine, and
    in the B-chain of SEQ ID NO: 4, Xaa8 is glutamic acid, Xaa9 is absent, Xaa10 is threonine, and Xaa11 is proline.
  11. The insulin analog peptide of claim 10, wherein the peptide comprises an amino acid sequence represented by SEQ ID NO: 22.
  12. A nucleic acid encoding the insulin analog peptide according to any of claims 1 to 11.
  13. The nucleic acid of claim 12, wherein the nucleic acid comprises a nucleotide sequence selected from the group consisting of SEQ ID NOS: 15, 17, 19, and 21.
  14. A recombinant expression vector comprising the nucleic acid according to claim 12.
  15. A transformant which is transformed with the recombinant expression vector according to claim 14.
  16. The transformant of claim 15, wherein the transformant is E. coli.
  17. A method of preparing the insulin analog peptide according to claim 1, comprising:
    a) preparing a recombinant expression vector comprising a nucleic acid encoding the insulin analog peptide according to claim 1;
    b) transforming the recombinant expression vector into a host cell and obtaining a transformant therefrom;
    c) culturing the transformant and expressing the insulin analog peptide; and
    d) isolating and purifying the expressed insulin analog peptide.
  18. The method of claim 17, wherein the nucleic acid comprises a nucleotide sequence selected from the group consisting of SEQ ID NOS: 15, 17, 19, and 21.
  19. The method of claim 17, wherein the transformant is E. coli.
  20. The method of claim 17, wherein the isolating and purifying comprise:
    d-1) obtaining the transformant cells from the culture and pulverizing the same;
    d-2) recovering the expressed insulin analog peptide from the pulverized cell lysate followed by refolding the same;
    d-3) purifying the refolded insulin analog peptide by cation exchange chromatography;
    d-4) treating the purified insulin analog peptide with trypsin and carboxypeptidase B; and
    d-5) sequentially purifying the treated insulin analog peptide by cation exchange chromatography and anion exchange chromatography.
  21. A pharmaceutical composition for treating diabetes comprising the insulin analog peptide according to any of claims 1 to 11 and a pharmaceutically acceptable carrier.
PCT/KR2016/009606 2015-08-28 2016-08-29 Novel insulin analogs and use thereof WO2017039267A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
JP2018511151A JP6976930B2 (en) 2015-08-28 2016-08-29 New insulin analogs and their uses
CA2999823A CA2999823A1 (en) 2015-08-28 2016-08-29 Novel insulin analogs and use thereof
CN201680062814.5A CN108350056B (en) 2015-08-28 2016-08-29 Novel insulin analogues and uses thereof
AU2016317449A AU2016317449B2 (en) 2015-08-28 2016-08-29 Novel insulin analogs and use thereof
EA201890584A EA201890584A1 (en) 2015-08-28 2016-08-29 NEW INSULIN ANALOGUES AND THEIR APPLICATION
EP16842233.5A EP3341404B1 (en) 2015-08-28 2016-08-29 Novel insulin analogs and use thereof
UAA201802993A UA125928C2 (en) 2015-08-28 2016-08-29 Novel insulin analogs and use thereof
ZA2018/02017A ZA201802017B (en) 2015-08-28 2018-03-27 Novel insulin analogs and use thereof
PH12018500690A PH12018500690A1 (en) 2015-08-28 2018-03-27 Novel insulin analogs and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20150121819 2015-08-28
KR10-2015-0121819 2015-08-28

Publications (1)

Publication Number Publication Date
WO2017039267A1 true WO2017039267A1 (en) 2017-03-09

Family

ID=58189026

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2016/009606 WO2017039267A1 (en) 2015-08-28 2016-08-29 Novel insulin analogs and use thereof

Country Status (15)

Country Link
US (2) US10017557B2 (en)
EP (1) EP3341404B1 (en)
JP (2) JP6976930B2 (en)
KR (1) KR20170026284A (en)
CN (2) CN108350056B (en)
AR (1) AR105822A1 (en)
AU (1) AU2016317449B2 (en)
CA (1) CA2999823A1 (en)
EA (1) EA201890584A1 (en)
PH (1) PH12018500690A1 (en)
TW (1) TWI781083B (en)
UA (1) UA125928C2 (en)
UY (1) UY36870A (en)
WO (1) WO2017039267A1 (en)
ZA (1) ZA201802017B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018143729A1 (en) 2017-02-03 2018-08-09 한미약품 주식회사 Conjugate of bioactive material having enhanced sustainability and use thereof
WO2020130749A1 (en) 2018-12-21 2020-06-25 한미약품 주식회사 Pharmaceutical composition comprising insulin and triple agonist having activity with respect to all of glucagon and glp-1 and gip receptor

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UY36870A (en) * 2015-08-28 2017-03-31 Hanmi Pharm Ind Co Ltd NEW INSULIN ANALOGS
WO2018056764A1 (en) 2016-09-23 2018-03-29 한미약품 주식회사 Insulin analog having reduced binding force to insulin receptor, and use thereof
WO2018105988A1 (en) 2016-12-05 2018-06-14 한미약품 주식회사 Conjugate having attenuated immune response
TWI798209B (en) 2017-03-23 2023-04-11 南韓商韓美藥品股份有限公司 A conjugate of insulin analog with reduced affinity to insulin receptor and use thereof
EP3900734A4 (en) 2018-12-21 2022-10-12 Hanmi Pharm. Co., Ltd. Pharmaceutical composition containing insulin and glucagon
CN111220604B (en) * 2019-12-06 2022-03-15 东北农业大学 Method for measuring total starch content in meat product
KR102663243B1 (en) 2021-08-30 2024-05-03 국립순천대학교산학협력단 Trypsin specific fluorescent probe and uses thereof
KR102574341B1 (en) 2021-08-30 2023-09-04 순천대학교 산학협력단 Amino acid specific masking agent and uses thereof
CN114805544B (en) * 2022-06-23 2022-09-09 北京惠之衡生物科技有限公司 Insulin lispro precursor, recombinant genetic engineering bacterium thereof and construction method thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2536040A1 (en) 1975-08-13 1977-02-24 Hoechst Ag Prolonged action insulin analogues - with A1-glycine replaced by D-alanine or D-serine
US5716927A (en) 1988-12-23 1998-02-10 Novo Nordisk A/S Insulin analogs having a modified B-chain
KR101324828B1 (en) * 2010-06-08 2013-11-01 한미사이언스 주식회사 An single chain-insulin analog complex using an immunoglobulin fragment
WO2014133324A1 (en) 2013-02-26 2014-09-04 한미약품 주식회사 Novel insulin analog and use thereof
WO2015108398A1 (en) 2014-01-20 2015-07-23 한미약품 주식회사 Long-acting insulin and use thereof

Family Cites Families (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5175145A (en) 1988-08-26 1992-12-29 Amylin Pharmaceuticals, Inc. Treatment of diabetes mellitus with amylin agonists
JPH04502465A (en) * 1988-12-23 1992-05-07 ノボ ノルディスク アクティーゼルスカブ human insulin analogue
US5422339A (en) 1991-03-19 1995-06-06 Joslin Diabetes Center, Inc. Peptides having insulin autoantibody but not insulin receptor binding capacity
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
DE69731289D1 (en) 1996-03-18 2004-11-25 Univ Texas IMMUNGLOBULIN-LIKE DOMAIN WITH INCREASED HALF-VALUE TIMES
DE19825447A1 (en) * 1998-06-06 1999-12-09 Hoechst Marion Roussel De Gmbh New insulin analogues with increased zinc formation
ATE284415T1 (en) * 1999-01-06 2004-12-15 Genentech Inc MUTATED VARIANT OF INSULIN-LIKE GROWTH FACTOR-I (IGF-I)
CN1635900A (en) 2001-08-28 2005-07-06 伊莱利利公司 Pre-mixes of GLP-1 and basal insulin
AR036711A1 (en) 2001-10-05 2004-09-29 Bayer Corp PEPTIDES THAT ACT AS GLON-RECEPTOR AGONISTS AND AS GLUCAGON RECEPTOR ANTAGONISTS AND THEIR PHARMACOLOGICAL USE METHODS
EP1448222A4 (en) 2001-10-19 2006-05-17 Lilly Co Eli Biphasic mixtures of glp-1 and insulin
DE10227232A1 (en) 2002-06-18 2004-01-15 Aventis Pharma Deutschland Gmbh Sour insulin preparations with improved stability
AU2004234345A1 (en) 2003-04-29 2004-11-11 Eli Lilly And Company Insulin analogs having protracted time action
SI1648933T1 (en) 2003-07-25 2010-01-29 Conjuchem Biotechnologies Inc Long lasting insulin derivatives and methods thereof
ES2372495T3 (en) 2003-11-13 2012-01-20 Hanmi Holdings Co., Ltd METHOD FOR MASS PRODUCTION OF THE CONSTANT REGION OF IMMUNOGLOBULIN.
JP5366546B2 (en) 2005-08-16 2013-12-11 ノボ・ノルデイスク・エー/エス Method for producing mature insulin polypeptide
PL2074141T3 (en) * 2006-09-22 2017-02-28 Novo Nordisk A/S Protease resistant insulin analogues
US7790677B2 (en) 2006-12-13 2010-09-07 Elona Biotechnologies Insulin production methods and pro-insulin constructs
JP2008169195A (en) 2007-01-05 2008-07-24 Hanmi Pharmaceutical Co Ltd Insulinotopic peptide drug combo using carrier material
EP2170945A1 (en) 2007-07-16 2010-04-07 Novo Nordisk A/S Protease stabilized, pegylated insulin analogues
EP2017288A1 (en) 2007-07-16 2009-01-21 Novo Nordisk A/S Protease stabilized, pegylated insulin analogues
JP2009019027A (en) 2007-07-16 2009-01-29 Hanmi Pharmaceutical Co Ltd Insulin secretion peptide derivative in which amino acid of amino terminal is varied
US8575096B2 (en) * 2007-08-13 2013-11-05 Novo Nordisk A/S Rapid acting insulin analogues
EP2708554A1 (en) 2007-08-15 2014-03-19 Novo Nordisk A/S Insulin analogues with an acyl and alkylene glycol moiety
CN101157725B (en) * 2007-10-24 2012-07-25 中国药科大学 Preparation method of human insulin analogue and usage thereof
CN107115523A (en) 2007-11-16 2017-09-01 诺沃—诺迪斯克有限公司 Pharmaceutical composition comprising the peptides of GLP 1 or exenatide and basal insulin peptide
BRPI0907371A2 (en) 2008-01-09 2015-11-24 Sanofi Aventis Deutschland insulin derivatives with a very delayed time-action profile
DE102008003568A1 (en) 2008-01-09 2009-07-16 Sanofi-Aventis Deutschland Gmbh New insulin analogs useful for treating diabetes
DE102008025008A1 (en) 2008-05-24 2009-11-26 Sanofi-Aventis Deutschland Gmbh Insulin analogs which comprise A chain and B chain with disulfide bonds for use in treatment of diabetes
JP5695909B2 (en) 2008-01-09 2015-04-08 サノフィ−アベンティス・ドイチュラント・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング Novel insulin derivatives with extremely delayed time action profiles
EP2254905B1 (en) * 2008-03-14 2016-12-14 Novo Nordisk A/S Protease-stabilized insulin analogues
EP2910570B1 (en) 2008-03-18 2016-10-12 Novo Nordisk A/S Protease stabilized, acylated insulin analogues
WO2009129250A2 (en) 2008-04-14 2009-10-22 Case Western Reserve University Meal-time insulin analogues of enhanced stability
MX2010011329A (en) * 2008-04-22 2011-03-15 Univ Case Western Reserve Isoform-specific insulin analogues.
HUE037449T2 (en) 2008-10-17 2018-08-28 Sanofi Aventis Deutschland Combination of an insulin and a glp-1 agonist
CN102256992B (en) 2008-12-19 2015-04-22 印第安纳大学研究及科技有限公司 Insulin analogs
JP5755566B2 (en) 2008-12-19 2015-07-29 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーションIndiana University Research And Technology Corporation Amide insulin prodrug
MA33221B1 (en) 2009-03-27 2012-04-02 Glaxo Group Ltd Integration of medicines and facilities
WO2011028813A2 (en) 2009-09-01 2011-03-10 Case Western Reserve University Insulin analogues of enhanced receptor-binding specificity
RU2537239C2 (en) 2009-11-13 2014-12-27 Санофи-Авентис Дойчланд Гмбх Pharmaceutical composition containing agonist glp-1, insulin and methionine
WO2011075393A2 (en) 2009-12-18 2011-06-23 Indiana University Research And Technology Corporation Glucagon/glp-1 receptor co-agonists
KR101058209B1 (en) 2009-12-30 2011-08-22 전자부품연구원 Fast Synchronization Method of Digital Broadcasting System
AR081066A1 (en) 2010-04-02 2012-06-06 Hanmi Holdings Co Ltd INSULIN CONJUGATE WHERE AN IMMUNOGLOBULIN FRAGMENT IS USED
KR101330868B1 (en) 2010-06-08 2013-11-18 한미사이언스 주식회사 Insulin derivative drug conjugate using an immunoglobulin fragment
EP2582719B1 (en) * 2010-06-16 2016-08-10 Indiana University Research and Technology Corporation Single chain insulin agonists exhibiting high activity at the insulin receptor
KR101382593B1 (en) 2010-07-21 2014-04-10 한미사이언스 주식회사 Novel long-acting glucagon conjugate and pharmaceutical composition comprising the same for the prevention and treatment of obesity
WO2012015692A2 (en) 2010-07-28 2012-02-02 Smartcells, Inc. Recombinantly expressed insulin polypeptides and uses thereof
KR20120013727A (en) 2010-08-06 2012-02-15 삼성전자주식회사 Display apparatus and control method thereof
EA201390796A1 (en) 2011-01-20 2014-07-30 Зилэнд Фарма А/С APPLICATION OF ACYLATED GLUCAGON ANALOG
WO2012116818A1 (en) 2011-03-02 2012-09-07 Oerlikon Trading Ag, Trübbach Sliding component coated with metal-comprising carbon layer for improving wear and friction behavior by tribological applications under lubricated conditions
CN102675452B (en) 2011-03-17 2015-09-16 重庆富进生物医药有限公司 Tool continues the conjugate of insulin human that is hypoglycemic and that combined by height and analogue
US9341445B2 (en) 2011-05-03 2016-05-17 Teijin Aramid Bv Antiballistic panel with first and second laminates having fibers of different tensile modulus
WO2012167251A1 (en) 2011-06-02 2012-12-06 Prolor Biotech Inc. Long-acting glp-1/glucagon receptor agonists
UA113626C2 (en) 2011-06-02 2017-02-27 A COMPOSITION FOR THE TREATMENT OF DIABETES CONTAINING THE DURABLE INSULIN CON conjugate AND THE DUAL ACTION INSULINOTROPIC PIPIDE
DK2718318T3 (en) 2011-06-10 2018-11-05 Hanmi Science Co Ltd New oxyntomodulin derivatives and pharmaceutical compositions for the treatment of obesity comprising these
WO2012173422A1 (en) 2011-06-17 2012-12-20 Hanmi Science Co., Ltd. A conjugate comprising oxyntomodulin and an immunoglobulin fragment, and use thereof
US9165768B2 (en) 2011-12-16 2015-10-20 Lg Innotek Co., Ltd. Method for deposition of silicon carbide and silicon carbide epitaxial wafer
EP2804621A4 (en) 2012-01-20 2015-11-18 Univ Case Western Reserve Glutamic acid-stabilized insulin analogues
CN104136626B (en) * 2012-03-01 2017-05-03 诺沃—诺迪斯克有限公司 N-terminally modified oligopeptides and uses thereo
KR101665009B1 (en) 2012-03-09 2016-10-11 한미사이언스 주식회사 Pharmaceutical composition for the prevention or treatment of non-alcoholic fatty liver disease
KR101968344B1 (en) 2012-07-25 2019-04-12 한미약품 주식회사 A composition for treating hyperlipidemia comprising oxyntomodulin analog
AR091902A1 (en) 2012-07-25 2015-03-11 Hanmi Pharm Ind Co Ltd LIQUID FORMULATION OF A PROLONGED INSULIN CONJUGATE
AR094821A1 (en) 2012-07-25 2015-09-02 Hanmi Pharm Ind Co Ltd LIQUID FORMULATION OF AN INSULINOTROPIC PEPTIDE CONJUGATE OF PROLONGED ACTION
AR092862A1 (en) 2012-07-25 2015-05-06 Hanmi Pharm Ind Co Ltd LIQUID FORMULATION OF PROLONGED ACTION INSULIN AND AN INSULINOTROPIC PEPTIDE AND PREPARATION METHOD
AR092873A1 (en) 2012-09-26 2015-05-06 Cadila Healthcare Ltd PEPTIDES AS TRIPLE AGONISTS OF GIP, GLP-1 AND GLUGAGON RECEPTORS
EA034499B1 (en) 2012-11-06 2020-02-13 Ханми Фарм. Ко., Лтд. Liquid formulation of protein conjugate comprising oxyntomodulin and an immunoglobulin fragment
MX361083B (en) * 2013-02-26 2018-11-27 Hanmi Pharma Co Ltd Site-specific insulin conjugate.
AR100639A1 (en) 2014-05-29 2016-10-19 Hanmi Pharm Ind Co Ltd COMPOSITION TO TREAT DIABETES THAT INCLUDES CONJUGATES OF PROLONGED INSULIN ANALOGS AND CONJUGATES OF PROLONGED INSULINOTROPIC PEPTIDES
KR101676542B1 (en) * 2014-12-30 2016-11-16 건국대학교 산학협력단 Use of proinsulin for immune system modulation
UY36870A (en) 2015-08-28 2017-03-31 Hanmi Pharm Ind Co Ltd NEW INSULIN ANALOGS

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2536040A1 (en) 1975-08-13 1977-02-24 Hoechst Ag Prolonged action insulin analogues - with A1-glycine replaced by D-alanine or D-serine
US5716927A (en) 1988-12-23 1998-02-10 Novo Nordisk A/S Insulin analogs having a modified B-chain
KR101324828B1 (en) * 2010-06-08 2013-11-01 한미사이언스 주식회사 An single chain-insulin analog complex using an immunoglobulin fragment
WO2014133324A1 (en) 2013-02-26 2014-09-04 한미약품 주식회사 Novel insulin analog and use thereof
WO2015108398A1 (en) 2014-01-20 2015-07-23 한미약품 주식회사 Long-acting insulin and use thereof

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
DATABASE Genbank [O] 1 June 2015 (2015-06-01), XP055367616, Database accession no. AKI70564.1 *
DATABASE Genbank [O] 13 May 2015 (2015-05-13), XP055367621, Database accession no. NM_001291897.1 *
DATABASE Genbank [O] 27 April 1993 (1993-04-27), XP055367612, Database accession no. AAA72172.1 *
JORGENSEN ET AL.: "Solution structure of the superactive monomeric Des- [Phe(B25)] human insulin mutant: elucidation of the structural basis for the monomerization of Des-[Phe(B25)] insulin and the dimerization of native insulin", JOURNAL OF MOLECULAR BIOLOGY, vol. 257, 1996, pages 684 - 699, XP055367638 *
KELLER ET AL.: "Flexibility and bioactivity of insulin: an NMR investigation of the solution structure and folding of an unusually flexible human insulin mutant with increased biological activity", BIOCHEMISTRY, vol. 40, 2001, pages 10732 - 10740, XP055367634 *
SCHEIT: "Nucleotide Analogs", 1980, JOHN WILEY
See also references of EP3341404A4
UHLMANPEYMAN, CHEMICAL REVIEWS, vol. 90, 1990, pages 543 - 584

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018143729A1 (en) 2017-02-03 2018-08-09 한미약품 주식회사 Conjugate of bioactive material having enhanced sustainability and use thereof
WO2020130749A1 (en) 2018-12-21 2020-06-25 한미약품 주식회사 Pharmaceutical composition comprising insulin and triple agonist having activity with respect to all of glucagon and glp-1 and gip receptor

Also Published As

Publication number Publication date
US10017557B2 (en) 2018-07-10
JP6976930B2 (en) 2021-12-08
EP3341404C0 (en) 2024-04-03
AR105822A1 (en) 2017-11-15
JP2018526001A (en) 2018-09-13
AU2016317449A1 (en) 2018-04-19
KR20170026284A (en) 2017-03-08
UY36870A (en) 2017-03-31
UA125928C2 (en) 2022-07-13
EP3341404B1 (en) 2024-04-03
AU2016317449B2 (en) 2021-01-07
CN108350056A (en) 2018-07-31
EP3341404A1 (en) 2018-07-04
TW201710288A (en) 2017-03-16
TWI781083B (en) 2022-10-21
ZA201802017B (en) 2019-01-30
JP2021168687A (en) 2021-10-28
US20170066811A1 (en) 2017-03-09
CN108350056B (en) 2021-09-21
JP7252280B2 (en) 2023-04-04
PH12018500690A1 (en) 2018-10-15
EA201890584A1 (en) 2018-08-31
US10647753B2 (en) 2020-05-12
EP3341404A4 (en) 2019-08-21
CA2999823A1 (en) 2017-03-09
US20180282388A1 (en) 2018-10-04
CN113637065A (en) 2021-11-12

Similar Documents

Publication Publication Date Title
AU2016317449B2 (en) Novel insulin analogs and use thereof
WO2018056764A1 (en) Insulin analog having reduced binding force to insulin receptor, and use thereof
WO2015108398A1 (en) Long-acting insulin and use thereof
WO2017052305A1 (en) Method of insulin production
WO2012169798A2 (en) Novel oxyntomodulin derivatives and pharmaceutical composition for treating obesity comprising the same
WO2016006963A1 (en) Insulin analogue
WO2014035179A1 (en) Mitochondrial targeting peptide
WO2019125059A1 (en) Therapeutic enzyme fusion protein having novel structure and use thereof
WO2019066586A1 (en) Long-acting conjugate of glucagon-like peptide-2 (glp-2) derivative
WO2019066570A1 (en) Long-acting single-chain insulin analog and conjugate thereof
WO2020130300A1 (en) Novel immunosuppressive interleukin 2
WO2018174668A2 (en) Insulin analog complex with reduced affinity for insulin receptor and use thereof
WO2018004294A9 (en) Pharmaceutical composition comprising mutant human growth hormone protein or transferrin fusion protein thereof as effective ingredient
WO2021107519A1 (en) Biotin moiety-conjugated polypeptide and pharmaceutical composition for oral administration comprising the same
WO2020251163A1 (en) Non-toxic protease having improved productivity
WO2009145573A9 (en) Recombinant escherichia coli for producing recombinant human variation interferon-beta proteins wherein methionine at the amino end is removed, and preparation method thereof
WO2019035672A1 (en) Acylated oxyntomodulin peptide analog
WO2023121328A1 (en) Novel immunosuppressive interleukin 2 analogue
WO2022245179A1 (en) Composition for combination therapy comprising growth differentiation factor-15 variant and glucagon-like peptide-1 receptor agonist
WO2020032423A1 (en) Method for preparation of active form of long-acting insulin analogue conjugate by using clostripain
WO2020017916A1 (en) Pharmaceutical composition comprising polypeptide
WO2020130594A1 (en) Fusion protein comprising human lefty a protein variants and use thereof
EA041658B1 (en) NEW INSULIN ANALOGUES AND THEIR APPLICATIONS
EA045778B1 (en) NEW INSULIN ANALOGUES AND THEIR APPLICATION

Legal Events

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

Ref document number: 16842233

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2018511151

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2999823

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: A201802993

Country of ref document: UA

Ref document number: 201890584

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 12018500690

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 11201802523X

Country of ref document: SG

Ref document number: 2016842233

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016317449

Country of ref document: AU

Date of ref document: 20160829

Kind code of ref document: A