WO2012115640A1 - Compositions liquides contenant de l'insuline et leurs procédés de fabrication - Google Patents

Compositions liquides contenant de l'insuline et leurs procédés de fabrication Download PDF

Info

Publication number
WO2012115640A1
WO2012115640A1 PCT/US2011/025928 US2011025928W WO2012115640A1 WO 2012115640 A1 WO2012115640 A1 WO 2012115640A1 US 2011025928 W US2011025928 W US 2011025928W WO 2012115640 A1 WO2012115640 A1 WO 2012115640A1
Authority
WO
WIPO (PCT)
Prior art keywords
insulin
proinsulin
sequence
human insulin
recombinant human
Prior art date
Application number
PCT/US2011/025928
Other languages
English (en)
Inventor
Ronald E. ZIMMERMAN
David John STOKELL
Michael Patrick AKERS
Original Assignee
Elona Biotechnologies
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 Elona Biotechnologies filed Critical Elona Biotechnologies
Priority to PCT/US2011/025928 priority Critical patent/WO2012115640A1/fr
Publication of WO2012115640A1 publication Critical patent/WO2012115640A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner

Definitions

  • the invention relates to liquid insulin compositions and methods for preparing liquid insulin compositions.
  • Insulin is a hormone that regulates glucose metabolism in animals.
  • Insulin is a polypeptide hormone secreted by beta-cells of the pancreas. This hormone is made up of two polypeptide chains, an A-chain of 21 amino acids, and a B-chain of 30 amino acids. These two chains are linked to one another in the mature form of the hormone by two interchain disulphide bridges. The A-chain also features one intra-chain disulphide bridge.
  • Insulin is a hormone that is synthesized in the body in the form of a single-chain precursor molecule, pro-insulin.
  • Pro-insulin is a molecule that from the N-terminus to the C-terminus has a pre-peptide of 24 amino acids, followed by the B-chain peptide, followed by the C-peptide of 35 amino acids, and then the A-chain peptide.
  • the C-peptide of this precursor insulin molecule contains the two amino acids, lysine-arginine (KR) at its carboxy end (where it attaches to the A-chain), and the two amino acids, arginine-arginine (RR) at its amino end (where it attaches to the B-chain).
  • KR lysine-arginine
  • RR arginine-arginine
  • the C-peptide is cleaved away, leaving the A-chain and the B-chain connected directly to one another in its active form.
  • a third method utilizes yeast, especially Saccharomyces cerevisiae, to secrete the insulin precursor into the medium (Thim, et al. (1986), P.N.A.S., USA., 83: 6766-6770).
  • Chance et al. reported a process for preparing insulin by producing each of the A and B chains of insulin in the form of a fusion protein by culturing E. coli that carries a vector compromising a DNA encoding the fusion protein, cleaving the fusion protein with cyanogen bromide to obtain the A and the B chains, sulfonating the A and B chains to obtain sulfonated chains, reacting the sulfonated B chain with an excess amount of the sulfonated A chain; and then purifying the resultant products to obtain insulin.
  • Frank et al. relates to a process for preparing insulin in the form of a fusion protein in E. coll
  • proinsulin is produced in the form of a fusion protein by culturing E. coli which carries a vector comprising a nucleic acid sequence (DNA) encoding for the fusion protein, cutting the fusion protein with cyanogen bromide to obtain proinsulin, sulfonating the proinsulin and separation of the sulfonated proinsulin, refolding the sulfonated proinsulin to form correct disulfide bonds, treating the refolded proinsulin with trypsin and carboxypeptidase B, and then purifying the resultant product to obtain insulin.
  • DNA nucleic acid sequence
  • the yield of the refolded proinsulin having correctly folded disulfide bonds is reported to sharply decrease as the concentration of the proinsulin increases. This is allegedly due to, at least among other reasons, misfolding of the protein, and some degree of polymerization being involved. Hence, the process entails the inconvenience of using laborious purification steps during the recovery of proinsulin and consequently any final insulin product.
  • Thim et al. reported a process for producing insulin in yeast, Saccharomyces cerevisiae. This process has the steps of producing a single chain insulin analog having a certain amino acid sequence by culturing Saccharomyces cerevisiae cells, and isolating insulin therefrom through the steps of: purification, enzyme reaction, acid hydrolysis and a second purification. This process, however, results in an unacceptably low yield of insulin.
  • Trypsin is a typical serine protease, and hydrolyses a protein or peptide at the carboxyl terminal of an arginine or lysine residue (Enzymes, pp. 261-262 (1979), ed. Dixon, M. & Webb, E. C. Longman Group Ltd., London).
  • This unwanted hydrolysis results in the unwanted Arg(Ao)-insulin byproduct, and typically constitutes about 10% of the reaction yield.
  • an additional purification step is required. The necessity of an additional purification step makes the process much more time consuming, and thus expensive, to use. Moreover, an additional loss of yield may be expected from the necessity of this additional purification step.
  • U.S. Pat. No. 7,087,408 also describes insulin precursors and insulin precursor analogs having a mini C-peptide comprising at least one aromatic amino acid residue.
  • cleavage of the mini C-peptide from the B chain may be enabled by cleavage at the natural Lys(B 29 ) amino acid residue in the B chain giving rise to a des-Thr(B 30 ) insulin precursor or analogs thereof.
  • proinsulin constructs that do not have a conserved terminal dibasic amino acid sequence of the C-peptide region.
  • US Pat. No. 6,777,207 (Kjeldsen et al.) relates to a novel proinsulin peptide construct containing a shortened C-peptide that includes the two terminal amino acids, glycine-arginine or glycine-lysine at the carboxyl terminal end that connects to the A-chain of the peptide.
  • the B-chain of the proinsulin construct described therein has a length of 29 amino acids, in contrast to the native 30 amino acid length of the native B-chain in human insulin.
  • yeast provides a relatively low insulin yield, due to the intrinsically low expression levels of a yeast system as compared to E. coli.
  • 5,457,066 describes treating human insulin precursor with trypsin and carboxypeptidase B in an aqueous medium containing about 0.1 to about 2 moles of metal ions (specifically nickel ions), per mole of human insulin precursor.
  • metal ions specifically nickel ions
  • the use of metal ions as described in this patent may lead to potential production problems, among other concerns.
  • the present invention provides a process for producing a highly purified liquid active pharmaceutical ingredient (API) comprising a recombinant human insulin having an amino acid sequence that is about 95% homologous with the amino acid sequence of native human insulin.
  • API liquid active pharmaceutical ingredient
  • These preparations have a greatly reduced amount of related contaminant substances.
  • These related contaminant substances comprising, for example, high molecular weight substances, such as non-monomeric forms of insulin (multimeric forms including dimeric forms, etc.), chemically modified insulin molecules (e.g., desamido insulin forms (A 21 desamino insulin products), carbamylated insulin forms, norvaline contaminants, isopropyl phe products (e.g., isopropyl form of phenylanaline), and others).
  • the preparations of recombinant human insulin of the present invention may be described as comprising 2% or less of a specific desamido form of insulin, A 21 desamino insulin, and 1% or less non-monomeric species of insulin (multimeric species of insulin).
  • multimeric species generally refers to the insulin molecule that comprises multiple insulin monomers, e.g., dimers, trimers, tetramers, hexamer, etc.
  • the multimeric species may be further described as high molecular weight insulin species.
  • One aspect of the present invention is related to a process for producing insulin comprising the steps of culturing E. coli cells under conditions suitable for expression of a modified proinsulin sequence having the formula
  • R) is a tag sequence containing one or more amino acids or R ⁇ is absent with an
  • (BrB 30 ) and (A 1 -A 21 ) comprise amino acid sequences of native human insulin;
  • R 2 , R 3 and R 5 are Arg
  • R4 is any amino acid other than Gly, Lys or Arg or is absent;
  • X is a sequence that comprises one or more amino acids or is absent, provided that X is not EAEALQVGQVELGGGPGAGSLQPLALEGSLQ (SEQ ID NO: 2) and X does not comprise a C-terminal Gly, Lys, or Arg when R4 is absent; and
  • R ⁇ 5 is a tag sequence containing one or more amino acids or Re is absent
  • the next step is to disrupt the cultured E. coli cells to provide a composition comprising inclusion bodies containing the modified proinsulin sequence.
  • the inclusion bodies are solubilized followed by folding of the modified proinsulin sequence to provide a proinsulin derivative peptide.
  • the proinsulin derivative peptide is purified in a metal affinity chromatography column.
  • the next steps include protecting a lys amino acid residue of the proinsulin derivative peptide with one or more protecting compounds, enzymatically cleaving said blocked proinsulin derivative peptide to remove a connecting peptide and provide an intermediate solution comprising an insulin intermediate, purifying said intermediate solution in a chromatography column, enzymatically cleaving said intermediate insulin to produce an intermediate insulin solution, and purifying said insulin solution in a chromatography column to yield the insulin.
  • [0027] in one aspect of the present invention is related to a process for producing insulin from the modified proinsulin of Formula I.
  • the modified proinsulin is folded to provide a proinsulin derivative peptide.
  • the proinsulin derivative peptide is purified in a metal affinity chromatography column.
  • the next steps include protecting a lys amino acid residue of the proinsulin derivative peptide with one or more protecting compounds, enzymatically cleaving said blocked proinsulin derivative peptide to remove a connecting peptide and provide an intermediate solution comprising an insulin intermediate, purifying said intermediate solution in a chromatography column, enzymatically cleaving said intermediate insulin to produce an intermediate insulin solution, and purifying said insulin solution in a chromatography column to yield insulin.
  • FIG. 1 is a vector map of plasmid pTrcHis2A (Kan) with a proinsulin gene insert.
  • FIG. 2 is a process flow scheme for the purification of insulin.
  • FIG. 3 shows a map of peptide fragments of recombinant human insulin produced according to the methods of the invention.
  • the peptide map is based on non-reducing conditions and the fragments were created with V8 protease.
  • FIG. 3 discloses "USP Reference Standard” Fragment Numbers I-IV as SEQ ID NOS 28-33, respectively, in order of appearance, and "R&D Insulin” Fragment Numbers I-IV as SEQ ID NOS 28-33, respectively, in order of appearance.
  • FIG. 4 shows a map of peptide fragments of recombinant human insulin produced according to the methods of the invention.
  • the peptide map is based on reducing conditions and the fragments were created with V8 protease.
  • FIG. 4 discloses "USP Reference Standard” Fragment Numbers I- VI as SEQ ID NOS 33, 28, 30, 29 and 31-32, respectively, in order of appearance, and "R&D Insulin” Fragment Numbers I- VI as SEQ ID NOS 33, 28, 30, 29 and 31- 32, respectively, in order of appearance.
  • FIG. 5 shows mass spectrometry data for recombinant human insulin.
  • FIG. 6 shows an SDS PAGE reduced gel comparing electrophoretic patterns of wild-type human insulin and recombinant human insulin.
  • FIG. 7 shows an SDS PAGE non-reduced gel comparing electrophoretic patterns of wild- type human insulin and recombinant human insulin.
  • FIG. 8 shows an electrophoretic gel where recombinant (R&D Insulin) and wild-type insulin (Insulin Human, USP) have been subject to isoelectric focusing. Lanes 1 and 5 show isoelectric point markers (pi 3-10), lane 2 shows wild-type human insulin, and lanes 3 and 4 show recombinant human insulin from two different lots.
  • FIG. 9 is a table showing the levels of contamination of related substances in recombinant human insulin (R&D insulin) versus wild-type human insulin (Insulin human USP standard) as determined by RP-HPLC.
  • FIG. 10 an analytical HPLC overlay of liquid insulin product by the method according to one aspect of the invention, with crystal insulin product.
  • FIG. 11 is a vector map of plasmid pJ201 : 11351 with a HTHPI insert.
  • FIG. 12A is a process flow scheme for the purification of insulin.
  • FIG. 12B is a conventional process flow scheme for the purification and crystallization of insulin.
  • FIG. 13 is table of stability data comparing liquid insulin, according to one aspect of the invention, with insulin prepared by Eli Lilly.
  • FIG. 14 is a graph of United States Pharmacopeial (USP) insulin assay on day 1 and FIG. 15 is a graph of USP insulin assay on day 3.
  • USP United States Pharmacopeial
  • FIG. 16 is the resulting data from an analytical HPLC run of material produced by the herein described method, using the USP, standardized related substance method.
  • the present invention provides novel methods for manufacturing liquid insulin and novel compositions of liquid insulin.
  • Insulin is a hormone that regulates glucose metabolism in animals.
  • the insulin amino acid sequence is well preserved across vertebrates.
  • insulin is a vertebrate insulin.
  • insulin is a mammalian insulin.
  • insulin is porcine, equine, ovine, bovine, or porcine insulin.
  • insulin is human insulin.
  • the present invention relates to a highly purified preparation of recombinant human insulin.
  • the purified preparation of recombinant human insulin may be described as comprising an API (Active Pharmaceutical Ingredient) of recombinant human insulin having less than 2%, less than 1%, less than 0.5%, or even less than 0.1 1%, contaminant by weight of the total recombinant insulin protein preparation by total weight.
  • API Active Pharmaceutical Ingredient
  • the purified preparation of recombinant human insulin may be described as comprising an API of recombinant human insulin having less than 1%, less than 0.7%, less than 0.5%, or even less than 0.4%, multimeric species of insulin by weight of the total recombinant insulin protein preparation by total weight.
  • a preparation of recombinant insulin that is in a substantially liquid form and that has not been through a crystallization process is provided.
  • the preparation may be further described as a human recombinant insulin preparation in a substantially liquid form.
  • insulin of the invention includes insulin analogs or variants, i.e., polypeptides having insulin activity and substantial amino acid sequence identity to wild-type insulin.
  • analog or variant insulin proteins include proteins having the biological activity of at least 10%, 20%, 50%, 70%, 80%, 90%, 95%, 99% or 100% in comparison to the biological activity of wild type insulin.
  • analog or variant insulin proteins include proteins having at least 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity with wild-type insulin.
  • sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino acid or nucleic acid sequence).
  • the determination of percent homology between two sequences can be accomplished using a mathematical algorithm.
  • a non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, (1990) Proc. Natl. Acad. Sci. USA. 87:2264- 68, modified as in Karlin and Altschul, (1993) Proc. Natl. Acad. Sci. USA. 90:5873-77. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al, (1990) J. Mol.
  • Gapped BLAST can be utilized as described in Altschul et al, (1997) Nucleic Acids Research. 25(17):3389-3402.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • the methods of producing insulin described herein utilize a modified or variant pro-insulin sequence.
  • the modified proinsulin sequence of the present invention has the formula
  • R ⁇ is a tag sequence containing one or more amino acids, preferably with a C- terminal Arg or Lys, or R ⁇ is absent with an Arg or Lys present prior to the start of the B chain;
  • (B ! -B 30 ) and (Ai-A 21 ) comprise amino acid sequences of native human insulin
  • R 2 , R 3 and R 5 are Arg
  • Pv4 is any amino acid other than Gly, Lys or Arg or is absent, preferably Ala;
  • X is a sequence that comprises one or more amino acids or is absent, provided that X is not EAEALQVGQVELGGGPGAGSLQPLALEGSLQ (SEQ ID NO: 2) and X does not comprise a C-terminal Gly, Lys, or Arg when R4 is absent; and
  • R ⁇ 5 is a tag sequence containing one or more amino acids, preferably with a N- terminal Arg or Lys, or R is absent.
  • a pre or post-peptide that may be a native pre-peptide or an N-terminal multiple His-tag sequence, or any other commercially available tag utilized for protein purification, e.g. DSBC, Sumo, Thioredein, T7, S tag, Flag Tag, HA tag, VS epitope, Pel B tag, Xpress epitope, GST, MBP, NusA, CBP, or GFP.
  • DSBC Sumo, Thioredein, T7, S tag, Flag Tag, HA tag, VS epitope, Pel B tag, Xpress epitope, GST, MBP, NusA, CBP, or GFP.
  • R ⁇ or 3 ⁇ 4 is present in Formula I. It is preferably that the terminal amino acid of the pre or post-peptide that connects to the B-chain or A-chain comprise Arg or Lys.
  • Native pre-peptide has the sequence of MALWMRLLPLLALLALWGPDPAAA (SEQ ID NO: 3).
  • the N-terminal multiple His-tagged proinsulin construct comprises a 6-histidine (SEQ ID NO: 4) N-terminal tag and may have the sequence of MHHHHHHGGR (SEQ ID NO: 5).
  • the modified proinsulin sequence may replace the native 24 amino acid pre-peptide with the 6-histidine (SEQ ID NO: 4) N-terminal tag sequence.
  • Ri and/or 3 ⁇ 4 may be a sequence of one or more amino acids, e.g., preferably from 1 to 30 and more preferably from 6 to 10.
  • Native insulin comprises an A-chain having the sequence GIVEQCCTSICSLYQLENYCN (SEQ ID NO: 6) and a B-chain having the sequence FVNQHLCGSHLVEALYLVCGERGFFYTPKT (SEQ ID NO: 7).
  • connecting peptide or "C- peptide” is meant the connecting moiety "C" of the B-C-A polypeptide sequence of a single chain proinsulin molecule.
  • the N-terminus of the C-peptide connects to C-terminus of the modified B-chain, e.g., position 30 of the B-chain, and the C- terminus of the C-peptide connects to N-terminus of the A-chain, e.g., position 1 of the A-chain.
  • the C-peptide may have a sequence of the formula:
  • R 2 , R 3 , R4, R 5 , and X have the same meaning as in Formula I.
  • X may be a sequence having up to 40 amino acids, preferably up to 35 amino acids or more preferably up to 30 amino acids.
  • the C-peptide sequences of the present invention may include:
  • Preferred modified proinsulin sequences of the present invention may include:
  • the invention is directed to formulations of liquid insulin that are not reconstituted from a lyophilized or crystallized preparation of recombinant insulin.
  • the liquid insulin composition is a human liquid insulin composition.
  • the human liquid insulin is recombinantly produced.
  • the liquid insulin composition of the invention includes zinc ions.
  • zinc ions may be added to the liquid insulin preparation as a zinc chloride solution.
  • the liquid insulin composition of the invention includes HC1.
  • the formulation contains meta cresol at about 3.15mg/ml and glycerol at about 16mg/ml.
  • Liquid insulin preparations made according to the methods of the invention have several advantages.
  • one such advantage is that they are essentially free of contaminants and/or byproducts associated with the processing of a recombinant human insulin preparation that has first been lyophilized and/or crystallized and then subsequently reconstituted into a liquid form.
  • the lyophilization and/or crystallization of a recombinant human insulin liquid preparation has been associated with several disadvantages, including a host of impurities, decreased efficiency in product yield, changes in solubility and the presence of degradation products.
  • the major impurities associated with the crystallization and drying is the formation of multimers which occurs during the crystallization process and end up in the final product, which in turn leads to a lower overall purity.
  • the crystallization process decreases the yield, as the crystallization can never be taken to completion and therefore non- crystallized material is lost when the supernatant is discarded.
  • the methods of preparing insulin described herein eliminate the crystallization and drying steps that other manufacturers use to prepare recombinant insulin. Eliminating these steps has no negative impact on the purity of the insulin produced, but has the added advantage of reducing the amount of inactive insulin multimers in the liquid insulin product of the invention, whereas insulin reconstituted from lyophilized and crystallized insulin is contaminated with inactive insulin multimers.
  • the methods of producing insulin described herein generally include the following steps: fermentation/expression, Inclusion body isolation, solubilization of inclusion bodies; refolding processing and transformation of proinsulin to insulin ; and purification of insulin.
  • FIG. 2 illustrates a flow chart of preferred processes steps in producing liquid insulin according to embodiments of the present invention.
  • FIG. 12A compares processes steps in producing liquid insulin according to embodiments of the present invention in comparison with FIG. 12B which shows the current cumbersome process used to produce crystalline insulin.
  • the recombinant expression system is a transformed E. coli culture.
  • the transformed E. coli has been prepared and qualified as a master cell bank (MCB) containing proinsulin expressing vectors.
  • the cells of the MCB may be vertebrate or invertebrate cells, such as prokaryote or eukaryote cells, and most preferably the cells may be mammalian, bacterial, insect, or yeast cells.
  • the cell is a bacterial cell and in a further embodiment, the bacteria is E. coli.
  • the cell is a yeast cell and in a further embodiment, the yeast cell is S. cerevisiae or S. pombe.
  • E. coli cells may be cultured and disrupted to provide a composition comprising inclusion bodies.
  • the inclusion bodies contain the modified proinsulin sequence.
  • the proinsulin expressed by cells of the MCB according to the method of the invention may be secreted from the cells and include a secretory sequence.
  • proinsulin expressed by cells of the MCB are not secreted from the cells, and thus do not include a secretory sequence.
  • the step of solubilizing inclusion bodies may involve adjusting the pH to achieve complete solubilization of the modified proinsulin sequences.
  • the inclusion bodies may be solubilized by adjusting the pH to at least 10.5, preferably from 10.5 to 12.5, preferably from 11.8-12.
  • the pH may be adjusted by adding an alkali hydroxide such as NaOH or KOH to the composition of inclusion bodies.
  • the step of solubilization may use one or more reducing agents and/or chaotropic agent.
  • Suitable reducing agents may include those selected from the group consisting of 2-mercaptoethanol, L-cysteine hydrochloride monohydrate, dithiothreitol, dithierythritol, and mixtures thereof.
  • Suitable chaotropic agents include those selected from the group consisting of urea, thiourea, lithium perchlorate or guanidine hydrochloride, and mixtures thereof.
  • the solubilized inclusion bodies may be mixed in a refolding buffer, such as glycine or sodium carbonate, at a pH of 7-12, preferably from 10-11, preferably from 10.5-11, to refold the modified proinsulin sequences to a proinsulin derivative peptide.
  • a refolding buffer such as glycine or sodium carbonate
  • the solution with refolded material should be pH adjusted to 7-9, preferable 7.8-8.2, with or without the addition of an alkaline salt, preferably sodium chloride to a final concentration of lOOmM to 1M final concentration, preferably 500mM to 1M, preferably 700mM, and may be filtered and loaded onto a column, such as an immobilized metal-ion affinity chromatography (IMAC) column.
  • IMAC immobilized metal-ion affinity chromatography
  • resins suitable for embodiments of the present invention include Nickle Sepharose 6 Fast Flow (GE Healthcare), Nickle NTA Agarose (GE Healthcare), Chelating
  • IMAC Immobilized metal ion affinity chromatography
  • IMAC Immobilized metal ion affinity chromatography
  • This technique works by allowing proteins with an affinity for metal ions to be retained in a column containing immobilized metal ions, such as cobalt, nickel, or copper for the purification of proteins or peptides containing 3 or more sequential histidine residues or peptides and, iron, zinc or gallium for the purification of phosphorylated proteins or peptides.
  • immobilized metal ions such as cobalt, nickel, or copper for the purification of proteins or peptides containing 3 or more sequential histidine residues or peptides and, iron, zinc or gallium for the purification of phosphorylated proteins or peptides.
  • Many naturally occurring proteins do not have an affinity for metal ions, therefore recombinant DNA technology can be used to introduce such a protein tag into the relevant gene.
  • Methods used to elute the protein of interest include changing the pH, or adding a competitive molecule
  • one or more of the amino acids may be protected to prevent side reactions and impurities.
  • the addition of a protecting group to insulin may be added prior to addition of trypsin.
  • protecting groups may be used to protect the lysine residue of the B-chain.
  • a preferred protecting group is citriconic anhydride.
  • citriconic anhydride is preferably used to block Lys(B 2 9) in the proinsulin pro-lys-thr-arg-arg (SEQ ID NO: 21) amino acid sequence, and thus reducing the formation of desthreonine insulin impurity.
  • the citriconic anhydride protecting group may reduce the formation of impurities such as desthreonine insulin and arg(B 31 )-insulin.
  • an excess molar ratio of citriconic anhydride to proinsulin may be used.
  • about 10 fold molar excess or more of citriconic anhydride to proinsulin may be suitable, and more preferably, about 20 fold molar excess or more.
  • There is no upper limit on the excess molar ratio and the molar ratio may be as high as about 200 fold or about 300 fold.
  • proinsulin is subject to concentration by tangential flow filtration or diafiltration.
  • Proinsulin derivative peptide, with the blocking groups may be enzymatically cleaved, preferred by subjecting the proinsulin derivative peptide to trypsin digestion.
  • embodiments of the present invention may use commercially available rat, bovine, porcine or human trypsins or other isoenzymes or derivatives or variants thereof, it is also possible to use the following enzymes: trypsin from Fusarium oxysporum and from Streptomyces (S. griseus, S. exfoliatus, S. erythraeus, S. fradiae and S.
  • tryptase occurs at pH from about 7 to 10, and more preferably from 8.0 to 8.2.
  • the trypsin digest is quenched by adding glacial acetic acid. While it is contemplated that other additives may be employed, acetic acid appears to be most preferred and stable for this purpose.
  • Trypsin is an enzyme that has specific cleavage activity at the terminal arginine residues, and to a lesser extent, lysine residues, of the C-peptide. In the transformation reaction, it is required that the terminal arginine or lysine residues of the C-peptide be removed. In native human proinsulin, when trypsin cleaves at the lysine in position 64, it will be unable to remove the arginine at position 65, due to the fact that it requires at least one residue on both sides of a cleavage site. What results is the production of an unwanted by-product, arg(A 0 )-insulin.
  • This by-product constitutes a small loss in yield and generates an undesired contaminant.
  • the arg(A 0 )- insulin byproduct is preferentially not formed. When formed is less than 10%, and more preferably is less than 0.3% of total byproducts from the trypsin transformation reaction may be arg(Ao). This is because the trypsin no longer acts to cleave at this particular site of the proinsulin derivative peptide.
  • the insulin intermediate is subjected to deblocking.
  • Citriconic anhydride deblocking occurs by permitting the insulin intermediate to be warmed to a temperature of 15°C to 25°C, more preferably 18°C to 20°C, and the pH is adjusted to 2.5 to 3.5, more preferably 2.8 to 3.0.
  • the intermediate solution is preferably purified in a chromatography column, such as an ion exchange chromatography column or reverse phase chromatography column.
  • the intermediate solution may be purified in a chromatography column by eluting the insulin analog using a buffer comprising n-propanol or acetonitrile.
  • the buffer may also further comprise sodium sulfate, sodium chloride, phosphoric acid or acetic acid.
  • the insulin intermediate is subject to a carboxypeptidase B digestion.
  • carboxypeptidase B digestion occurs after the insulin intermediate purification step.
  • a trypsin inhibitor is added to the insulin prior to addition of carboxypeptidase B. Trypsin inhibitor is added in an amount that is equivalent to the trypsin added for the trypsin digest step.
  • a glycine solution is added to the insulin intermediate prior to addition of carboxypeptidase B.
  • glycine is added to adjust the pH of the insulin intermediate solution to 7-10, preferably 9.6 ⁇ 0.1.
  • the target concentration of glycine is 50mM using a 1M glycine stock.
  • the carboxypeptidase B is permitted to digest for at least 1- 16 hours, preferably at least 8 hours. A minimum of 10 hours is typically required, but overdigestion is not possible so there is no maximum time limit.
  • insulin is further purified after digestion by carboxypeptidase B.
  • carboxypeptidase B insulin is subject to purification via reverse phase chromatography.
  • the intermediate solution may be purified in a chromatography column by eluting the insulin using a buffer comprising an alcohol or organic solvent, preferably n-propanol or acetonitrile.
  • the buffer may also further comprise an alkali metal salt, preferable sodium sulfate.
  • the buffer may also further comprise an organic acid, suchas phosphoric acid.
  • insulin is concentrated and buffer exchanged using tangential flow filtration or diafiltration after reverse phase chromatography.
  • This step has been modified, in some embodiments, to dilute the reverse phase material down with (4X) cold lOOmM phosphate buffer at pH 7-8 prior to tangential flow filtration or diafiltration.
  • This is followed by tangential flow filtration or diafiltration/ into cold purified water.
  • the pH is maintained at 7.0- 9.0, preferably 7.5-8 during the exchanges.
  • the manufacturing process described herein results in a preparation of insulin in liquid active pharmaceutical ingredient (API) form.
  • API active pharmaceutical ingredient
  • the process eliminates the need to prepare a crystallized insulin that is later reconstituted.
  • the amount of inactive insulin multimers present in the liquid formulation is reduced in comparison to the amounts otherwise present in crystallized forms of insulin and reconstituted crystallized insulin.
  • the preparations comprise a pharmaceutically acceptable preparation comprising recombinant insulin and being essentially free of modified proinsulin sequences.
  • Formulations that are suitable for liquid insulin of the present invention may be used with a liquid API or by changing the API to one of the following:
  • Insulin mixes- regular insulin mixed with isophane insulin at specific ratios such as 70/30 or 50/50. These are currently sold as NPH (Neutral Protamine Hagedorn) insulin, such as Humulin N, Novolin N, Novolin NPH, and NPH Lletin II.
  • NPH Neutral Protamine Hagedorn
  • Lente formulations The insulin API prepared for regular insulin is pH controlled to get solubility of the Zinc-insulin complex. If the pH is not controlled a suspension of insulin crystals are formed. The size of these crystals is controlled to make either Lente or Ultra- lente formulations. These formulations are long acting and the Ultra-lente can act for longer than 24 hrs. Lente formulations are designed for single daily injections.
  • Inhaled insulins The insulin is spray dried instead of crystallized to form very small particles which can be sprayed into the lungs.
  • the large surface area sue to the small particles may allow for better absorption across the pulmonary cells into the blood stream.
  • Oral insulin- Spray dried insulin is formulated into a tablet.
  • the tablet would contain excipients that would allow it to hold together.
  • Insulin/insulin analog mixes- Mixing regular insulin with insulin analogs to create a fast acting to intermediate acting form (reg. insulin and Lis-pro insulin) or mixing regular insulin with a long acting analog to produce a medium to long acting form (reg. insulin and Lantus®).
  • the present example is provided to demonstrate the utility of the present invention for providing stable transformed E. coli that are capable of expressing recombinant human proinsulin protein.
  • the present example provides a description of the process to be followed to create a stable Master Cell Bank (MCB) containing recombinant E. coli cells capable of expressing recombinant human proinsulin.
  • MBC Master Cell Bank
  • Step 1 Construction of a purified proinsulin gene segment for insertion into the vector.
  • the initial gene construct was synthesized in a basic cloning vector, p J201 : 11351 vector (FIG. 11 ; SEQ ID NO: 34)
  • the gene construct included the N-terminal histidine tag, MHHHHHHGGR (SEQ ID NO: 5), modified B-chain, and modified C-peptide with the alanine codon in place of the native lysine and having the amino acid sequence
  • Step 2 Generation of the pTrcHis2A(Kan) vector containing proinsulin.
  • Commercially available pTrcHis2A(Kan) vector was modified to include a Kanamycin resistance gene in the middle of the Ampicillin resistance gene to negate the Ampicillin resistance prior to insertion of the proinsulin sequence into the vector.
  • Ampicillin resistance heightens the potential for allergic reactions to preparations made using vector constructs that include the Ampicillin resistance gene. Therefore it is preferable to eliminate the Ampicillin resistance in the constructs that are prepared and used.
  • the proinsulin gene was isolated from the DNA 2.0 plasmid using Ndel to cleave at the N-terminal side of the gene and EcoRl to cleave at the C-terminal side of the gene.
  • the Digested DNA was run over a 2% agarose gel to separate the plasmid DNA from the proinsulin gene.
  • a QIAquickTM (Qiagen) gel purification kit was then used to purify the gene construct.
  • Step 3 Transformation.
  • One microliter of the ligation reaction was used to transform competent E. coli cells BL21 with the pTrcHis2A( an) plasmid containing the proinsulin gene.
  • the transformed E. coli BL21 cells were plated on LB- an agar plates and incubated overnight at 37°C. Several clones were selected and sequenced. Clones with the correct sequence were then screened for expression.
  • the resulting clone is referred to as the His Tagged proinsulin pTrcHis2A(Kan) vector.
  • Step 4 Preparation of the Master Cell Bank (MCB).
  • MCB Master Cell Bank
  • sterile growth medium was inoculated with the recombinant BL21 E. coli containing the His Tagged proinsulin /pTrcHis2A(Kan) vector and incubated to allow cell growth.
  • the cells were harvested in an IS05 (class 100) environment under a biosafety cabinet and via centrifugation. Sterile medium and glycerol were added to cells. 1 mL aliquots of the cells were then dispensed into sterile ampoules and stored at -80°C. Aseptic techniques were utilized to generate the MCB.
  • the present example demonstrates the utility of the present invention as a method of providing a high yield, highly purified (reduced contaminant insulin related compounds) recombinant human insulin preparation from the pro-insulin expressing transformed E. coli (MCB) described in Example 1.
  • Step 1 Culturing of E. coli transformed with modified proinsulin sequence from the MCB of Example 1.
  • yeastolate purchased from VWR, Prod. # 90004-426 or - 488)
  • select phytone sodium chloride
  • purified water purified water
  • sterile Kanamycin solution sterile Kanamycin solution
  • Step 2 Disruption— Cells containing inclusion bodies expressing modified proinsulin are lysed in a basic Tris/salt buffer, using a Niro Soavi homogenizer (1100-1200 bar).
  • Step 3 Inclusion Body Washing— Contaminant protein removal is accomplished via two sequential washes with a Tris/Triton X-100 buffer, followed by two sequential washes with a Tris/Tween-20 buffer, and finally a single wash with a Tris/NaCl buffer.
  • Step 4 - Solubilization Inclusion bodies enriched with the modified proinsulin peptide are solubilized in 4-8M urea, preferably 6-8M urea containing reducing agents (2- mercaptoethanol, L-cysteine hydrochloride monohydrate). Complete solubilization is achieved by adjusting the pH to 10.5-12, preferably 11.8-12 with NaOH.
  • Step 5 Dilution refolding— The solubilized protein is then diluted into refolding buffer (20 mM Glycine, pH 10-11 at 6-10°C.) to a final concentration of 1 mg/ml and permitted to refold for 24 to 72 hours, preferentially about 48 hours, at 6-10°C. Higher protein concentration may be used in the refold if desired, however, overall refold efficiency will Sodium Chloride and Phosphate are then added to final concentrations of 700 mM and 25 mM respectively, followed by pH adjustment to 7.0 to 9.0, preferably 7.9-8.0 with 6M HC1.
  • Step 6 IMAC Chromatography— The dilute proinsulin derivative is loaded onto an IMAC column to a maximum capacity of ⁇ 26.5 mg main peak protein per ml of resin. A 75mM imidizole buffer is used to isocratically strip the majority of impurities from the column. The tagged proinsulin is then eluted isocratically using ⁇ 300 mM imidizole.
  • Step 7 Citriconic anhydride(CA) Blocking— To the IMAC pool, add citriconic anhydride at a molar ratio of 20:1 (CA to Pro-Insulin), while stirring at 4-10°C. Allow the sample to stir for not less than 3 hour at 4-10°C.
  • Step 8 Buffer exchange— To the IMAC main peak pool material, add EDTA to a final concentration of 20 mM. Exchange the buffer using a membrane with a suitable molecular weight cutoff (e.g. 3000 Da). The final buffer should be at least 97% exchanged to a 20 mM Tris-Cl, pH 7.0-10.0, preferably 8.1 at 8-10°C. A protein concentration of approximately 5 mg/ml is desirable.
  • Step 9 Trypsin Enzymatic Transformation/Proteolysis—
  • the buffer exchanged sample is digested with a 1500:1 mass ratio of main peak protein to trypsin, in the presence of 5mM CaCl.
  • the ratio of trypsin may be increased or decreased depending on the desired length of time for the reaction.
  • the digest is then quenched by the addition of acetic acid to > 700 mM.
  • Step 10 Citriconic anhydride Deblocking- The trypsin digest solution is then warmed to 18 to 20°C and the pH is adjusted to 2.8 to 3.0. The digest was stored at room temperature for not less than 10 hours to permit release of the citriconic anhydride.
  • the resulting preparation from Step 10 includes the di-Arg recombinant human insulin and other products resulting from the trypsin enzymatic digestion of the proinsulin sequence. This preparation is then subjected to the purification steps provided in Example 3 to provide a purified preparation of the recombinant human insulin product.
  • the C-peptide and tag (His-tag) have been dissociated from the recombinant human insulin sequence.
  • Step 11 Ion Exchange Chromatography— The digested material is loaded onto a cation exchange column and eluted with a NaCl gradient, in the presence of 20% n-propanol or acetonitrile at pH 2-5, preferably 4.0. RP-HPLC is used to pool the appropriate fractions containing the di-Arg recombinant human insulin peak of interest at the desired purity level.
  • Step 12 Reverse Phase Chromatography— The S-column pool containing the insulin is loaded onto an RPC30 or CI 8 reverse phase column and eluted using an n-propanol or acetonitrile gradient in the presence of 200mM sodium sulfate and 0.136% phosphoric acid. Fractions are immediately diluted 1 :4 with lOOmM Phosphate, pH 7-9, preferably 7.5-8 if n- propanol is used for elution; or 1:2 with lOOmM Phosphate, pH 7-9, preferably 7.5-8 if acetonitrile is use for elution or no dilution if acetonitrile is used for elution. RP-HPLC is used to pool the appropriate fractions containing the insulin peak of interest at the desired purity level.
  • Step 13 - Buffer Exchange Exchange the sample into WFI (water for injection) using a membrane with a suitable molecular weight cutoff (e.g. 3000 Da).
  • the pH of the solution should be monitored and maintained at 7.0-9.0, preferably 7.5-8.0.
  • the final sample is concentrated to 5-8 mg/ml, preferably 5-5.8mg/ml, with an adjusted pH of 7.0-9.0, preferably 7.5-8.0 at 6-10°C.
  • This material represents the liquid API form of the presently disclosed preparations of insulin.
  • the Insulin purified by Example 3 is formulated by diluting the API material with cold WFI to a final concentration of 4.3375 mg/ml.
  • a concentrated formulation buffer stock containing 80 mg/ml glycerol, 15.75 mg/ml meta cresol, 0.0985 mg/ml zinc chloride at pH 7.5 ⁇ 0.1 is added to the API material in a 1/5 ratio of formulation buffer stock to API.
  • the solution is mixed, followed by sterile filtration into appropriate vials in 10 ml aliquots.
  • Mass spectrometry was performed to determine the amino acid sequence, peptide map and disulfide bonds of the recombinant human insulin protein.
  • the reduced and non-reduced peptide mapping performed using Staph Aureus V8 protease showed the expected cleavages. These cleavages are shown in FIGS. 3 and 4. There are 4 cleavages in the non-reduced peptide map and 6 cleavages in the reduced peptide map. All of the recombinant human insulin protein fragments are identical to wild-type human insulin.
  • the molecular weight of recombinant human insulin was determined to be 5806 Da by mass spectrometry. This is within 2 Daltons of the theoretical mass of wild-type human insulin of 5807.58 Da.
  • the mass spectrographic data is shown in FIG. 5.
  • the eletrophoretic patterns of recombinant insulin and wild-type insulin were determined by polyacrylamide gel electrophoresis (PAGE) under non-reduced and reduced conditions. The results of these assays are shown in FIGS. 6 (reduced) and 7 (non reduced). In both cases, the electrophoretic patterns of recombinant human insulin and wild-type human insulin were identical. In each case, the proteins were run on NuPage 4-12% Bis-Tris gels with a MES SDS running buffer.
  • the isoelectric point of recombinant human insulin was determined by isoelectric focusing (IEF) gel electrophoresis.
  • the isoelectric point was identical to that of wild-type human insulin.
  • the gel can be seen in FIG. 8.
  • FIG. 9 is a summary of the identity of substances identified via HPLC which include insulin, A5/B4 desamido, A21 desamido, and insulin multimers. As shown in FIG. 9, the overall amount of related substances in the recombinant human insulin is lower than that of the insulin standard, indicating that the method of production described herein produces a lower amount of contaminants than found in insulin produced by other methods.
  • the method described herein produces recombinant human insulin of greater than 99% purity, while as shown in Figure 12, the standard had only 98.35% insulin.
  • recombinant insulin produced according to the method of the invention is shown in FIG. 9 to have 0.11% A 5 /B 4 desamido, while the standard has 0.23%, which is more than twice the contaminant level of the recombinant insulin made according to a method of the invention.
  • recombinant insulin produced according to the method of the invention is shown in FIG. 9 to have 0.10% A 21 desamido, while the standard has 0.49%, which is almost 5 times the level of contamination found in the recombinant insulin made according to the methods described herein.
  • the present example is provided to demonstrate the utility of the present compositions and methods for providing a liquid insulin product and method for manufacturing a liquid insulin product having a higher purity level than crystalline preparations of recombinant insulin.
  • the crystalline API insulin product employed in the present example was prepared by incubating a ⁇ 5mg/ml (insulin) sample at 18-21 °C overnight in the presence of 1.2M sodium chloride, 0.05M citric acid and 3mM zinc Chloride at pH 6.3. Crystals formed overnight and were harvested the following day via centrifugations, followed by drying in a vacuum dessicator to a final moisture content of 6-15%. For analytical analysis, crystals were reconstituted in 1 OmM hydrochloric acid.
  • FIG. 10 shows a comparison of liquid recombinant human insulin API prepared according to the methods provided herein and a crystalline recombinant human insulin API. As demonstrated in Figure 10, the liquid API is shown to contain 0.09% high molecular weight impurities compared with the crystal API which contains 0.34% high molecular weight impurities.
  • FIG. 13 shows stability data time points at 5°C, 25°C, and 40°C, over a 182 day period. The values are given in percent loss of main peak. The data demonstrates that the insulin produced by the herein described method(A) degrades at an equivalent rate to that of the currently marketed material(B), at all three temperatures.
  • the present example is provided to demonstrate the utility of the present invention for providing a product that provides an effective preparation for maintaining glucose levels in vivo.
  • a rabbit model was employed in the present study, and demonstrates the effectiveness of the present preparations for regulating glucose levels in all animals and in humans.
  • FIG. 14 and FIG. 15 shows the results of an in vivo animal (rabbit) study, looking at blood glucose levels pre and post subcutaneous injection of insulin produced by the herein described method compared with currently marketed insulin at days 1 and days 3.
  • the method was based on the current International Conference on Harmonisation (ICH) Harmonised Tripartite Guidelines, the United States Pharmacopeia guidelines for insulin assay.
  • Assay controls, vehicles and test articles and preparations information are summarized in the following Table 1.
  • the first treatment was administered on Day 1, and the second treatment was administered on Day 3.
  • control(saline), positive control (Humulin R), and liquid recombinant human insulin prepared according to the methods provided herein (inventive Insulin) were administered once on Day 1 and/or 3 during the study via subcutaneous injection.
  • the groups received a dose of positive control or test material on Day 1 , followed by a dose of positive control or test material on Day 3.
  • the dose levels for treated groups were 0.35 or 0.7 international units (IU) of inventive insulin and Humulin R, with combinations described in the above table.
  • the dose volume was maintained at 0.35mL/dose of test material, while the control group received 0.35mL of saline.
  • TABLES 2 and 3 summarize the average of six animals glucose values for test and control groups, which is show in Figures 14 and 15.
  • the present example demonstrates the utility of the present invention for providing a stable transformed E. coli master cell bank suitable for the commercial manufacture of high grade recombinant human insulin.
  • the analysis was performed to establish the qualification of the cell bank as a cGMP quality cell bank stock suitable for producing clinical grade human recombinant insulin. Plasmid copy number analysis was performed by qPCR using Beckman Coulter Genomic assays ECOAPH v 1.0 (detects the kanamycin resistance gene from transposon.
  • the Master cell bank was further analyzed to identify specific characteristics that define the stably transformed E. coli cells that carry the recombinant human insulin sequence containing plasmids. Some of the characteristics that may be used to define the transformed E. coli cells include plasmid copy number, DNA sequence analysis of isolated plasmids, genetic stability testing assessment, marker retention, cell viability count, and restriction mapping characterization. Plasmid DNA sequencing, plasmid copy number determination, and genetic stability testing assessment was conducted on transformed E. coli prepared according to these procedures described herein. The test results are summarized in Table 4.
  • Plasmid DNA was isolated from an LB broth plus kanamycin culture grown from an aliquote of each test article. Plasmid DNA was prepared from each culture using a Qiagen QIAmp DNA Mini kit, then assesed by agarose gel electropooresis and quantitated by spectrophotometry. The plasmid DNA was used as the template for DNA sequencing. The plasmid DNA was used as the template for DNA sequencing. The sequencing primers used are shown below:
  • DNA sequencing was performed via the BigDye® Terminator Cycle Sequencing Kit (Applied Biosystems). Sequencing reactions were purified then analyzed on an ABI PRISM 3730x1 DNA Analyzer. The raw data was analyzed using Sequencing Analysis software (Applied Biosystems). Sequence data was assembled and analyzed using the Sequencer software (Gene Codes).
  • PCR amplification of the test articles produced amplicons of the expected sizes for each primer set. No differences were observed in the derived consensus sequences generated for either test article and the reference sequences employed in this analysis.
  • Copy number analysis was performed by qPCR using the Beckman Coulter Genomics assays ECOAPH vl .O (detects the kanamycin resistance gene from transposon Tn903) and ECODNAP vl .l . (detects the E. coli DNA polymerase gene).
  • the ECODNAP vl . l assay was used as an endogenous control to normalize for the number of cells assayed.
  • a series of dilutions of the pTrcHis2 AKan plasmid were used to generate a standard curve to calibrate the ECOAPH vl.O target assay.
  • Total DNA extracted from the host E. coli cells was used to generate a standard curve to calibrate the ECODNAP vl .l assay. The assumptions were made that there is a single DNA polymerase gene.
  • the reactions were thermal cycled using the following conditions: 50°C for 2 minutes, 95°C for 10 minutes, followed by 40 cycles of 95°C for 15 seconds and 60°C for 1 minute. Data was collected by the ABI Prism 7900TM Sequence Detection System software (Applied Biosystems). Copy number was calculated as the number of copies (target gene) per cell (normalizing gene).
  • Method Bacterial Species Characterization: The Master Cell Bank samples were streaked on agar plates for colony isolation and incubated at 37C for approximately 16 hours. BL-21 Escherichia coli cells were processed in parallel to serve as a control. A single colony from each plate was transferred to a 0.85% solution, and the suspension used to inoculate API 20E kit test strips (bioMerieux) which are composed of 23 microtubes to perform 23 biochemical tests for the identification of glucose-fermenting Gram negative rods. The strips were incubated for 18-24 hours at 37°C then scored to identify the genus and species of the bacterium. Gram staining was performed from colonies representing both test articles and the control cells then fixed to glass slides. Each group of cells was Gram stained and viewed under lOOx magnification. E. coli cells were identified as rod shaped bacteria. Confirmation of the host control cells validated the assay and thus no repeat was necessary.
  • Results Bacterial Species Characterization: Gram stain results indicated the presence of gram negative cells. E03-INhis was identified to be Escherichia coli (99.9%ID). Gram stain results indicated the presence of gram negative cells.
  • Method Cell Purity Assessment: Three vials were selected fromthe master cell bank (E03-NhGH vials 44, 57, and 66). Six 100mm Tryptic Soy Agar plates were inoculated from each vial with 100 uL. Two additional plates were inoculated with PBS to serve as controls. Plates were incubated at 25 °C or 37°C for 7 days and monitored daily for heterogeneous growth.
  • Results Cell Purity Assessment: E03INhis displayed completely homogeneous lawn growth. Not growth was detectable on either negative control plate inoculated with PBS.
  • Method Phage Contamination Assessment: Supernatants were collected from both chloroform treated and non-treated MCB samples. The supernatants were plated with JM109 cells to test for plaque formation. Supernatants from K-12 and lambda phage were used as positive controls, and supernatant from phage-free XL 1 -Blue and lambda suspension medium were used as negative controls. Plates were all observed after 16 hours for plaque formation, and the number of plaques recorded.
  • Viable Cell counting was performed by preparing a series of dilutions from MCB E03INhis samples and plating three aliquots of each dilution on separate 100 mm LB agar plus kanamycin plates. As a negative control, 100 ⁇ of PBS was spread onto a 100 mm LB agar plus kanamycin plate. The plates were incubated at 37°C for approximately 16 hours. After incubation, the number of colonies was counted on the plates where individual colonies were observed. The viable cell count per milliliter of sample was calculated.
  • Figure 16 shows material produced by the current method analyzed using the related substances standardized United States Pharmacoeial (USP) method.
  • USP United States Pharmacoeial

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Dermatology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne de nouvelles préparations améliorées et des procédés pour la fabrication de préparations sensiblement liquides d'ingrédients pharmaceutiques actifs (IPA) à base d'insuline humaine recombinante. Les préparations liquides d'ingrédients pharmaceutiques actifs (IPA) à base d'insuline humaine recombinante purifiées sont sensiblement exemptes de sous-produits associés à la lyophilisation et/ou la cristallisation. Les procédés pour la fabrication de préparations sensiblement liquides d'ingrédients pharmaceutiques actifs (IPA) à base d'insuline humaine recombinante comprennent des étapes facultatives pour soumettre la préparation d'insuline recombinante à une lyophilisation et/ou une cristallisation. La présente invention permet ainsi d'obtenir un rendement amélioré d'insuline recombinante de plus grande pureté. L'invention concerne également des formulations hautement purifiées d'insuline humaine recombinante des préparations d'ingrédients pharmaceutiques actifs (IPA) à base d'insuline selon l'invention. L'invention concerne également des banques de cellules E. coli (MCB) transformées de manière stable capable d'exprimer l'insuline humaine recombinante.
PCT/US2011/025928 2011-02-23 2011-02-23 Compositions liquides contenant de l'insuline et leurs procédés de fabrication WO2012115640A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2011/025928 WO2012115640A1 (fr) 2011-02-23 2011-02-23 Compositions liquides contenant de l'insuline et leurs procédés de fabrication

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2011/025928 WO2012115640A1 (fr) 2011-02-23 2011-02-23 Compositions liquides contenant de l'insuline et leurs procédés de fabrication

Publications (1)

Publication Number Publication Date
WO2012115640A1 true WO2012115640A1 (fr) 2012-08-30

Family

ID=44625849

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/025928 WO2012115640A1 (fr) 2011-02-23 2011-02-23 Compositions liquides contenant de l'insuline et leurs procédés de fabrication

Country Status (1)

Country Link
WO (1) WO2012115640A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014084921A1 (fr) * 2012-11-27 2014-06-05 Rose Stuart Procédé de fabrication pharmaceutique et compositions
WO2014096985A3 (fr) * 2012-12-19 2015-07-16 Wockhardt Limited Composition aqueuse stable comprenant de l'insuline humaine ou un analogue ou un dérivé de celle-ci
CN113226284A (zh) * 2018-09-25 2021-08-06 美药星制药股份有限公司 高度纯化的重组人胰岛素(rhi)api及其生产方法

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5457066A (en) 1986-10-14 1995-10-10 Eli Lilly And Company Process for transforming a human insulin precursor to human insulin
US5962267A (en) 1995-02-15 1999-10-05 Hanil Synthetic Fiber Co., Ltd. Proinsulin derivative and process for producing human insulin
EP1338651A1 (fr) * 2000-12-01 2003-08-27 Yamanouchi Pharmaceutical Co. Ltd. Procede de depistage d'un remede
US6777207B2 (en) 1999-12-29 2004-08-17 Novo Nordisk A/S Method for making insulin precursors and insulin precursor analogues having improved fermentation yield in yeast
US7087408B2 (en) 1999-12-29 2006-08-08 Novo Nordisk A/S Method for making insulin precursors and insulin precursor analogs
WO2008016729A1 (fr) * 2006-08-04 2008-02-07 Nastech Pharmaceutical Company Inc. Compositions pour administration intranasale d'insuline humaine et leurs utilisations

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5457066A (en) 1986-10-14 1995-10-10 Eli Lilly And Company Process for transforming a human insulin precursor to human insulin
US5962267A (en) 1995-02-15 1999-10-05 Hanil Synthetic Fiber Co., Ltd. Proinsulin derivative and process for producing human insulin
US6777207B2 (en) 1999-12-29 2004-08-17 Novo Nordisk A/S Method for making insulin precursors and insulin precursor analogues having improved fermentation yield in yeast
US7087408B2 (en) 1999-12-29 2006-08-08 Novo Nordisk A/S Method for making insulin precursors and insulin precursor analogs
EP1338651A1 (fr) * 2000-12-01 2003-08-27 Yamanouchi Pharmaceutical Co. Ltd. Procede de depistage d'un remede
WO2008016729A1 (fr) * 2006-08-04 2008-02-07 Nastech Pharmaceutical Company Inc. Compositions pour administration intranasale d'insuline humaine et leurs utilisations

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Enzymes", 1979, LONGMAN GROUP LTD., pages: 261 - 262
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
ALTSCHUL ET AL., NUCLEIC ACIDS RESEARCH, vol. 25, no. 17, 1997, pages 3389 - 3402
CHAN ET AL., P.N.A.S., USA, vol. 78, 1981, pages 5401 - 5404
CHANG ET AL., BIOCHEM. J., vol. 329, 1998, pages 631 - 635
EMEA: "Scientific Discussion - NovoRapid", 2004, pages 1 - 16, XP002671681, Retrieved from the Internet <URL:http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human> [retrieved on 20120314] *
FRANK ET AL.: "Peptides: Proceedings of the 7th American Peptide Chemistry Symposium", 1981, PIERCE CHEMICAL COMPANY, pages: 721 - 728,729-
KARLIN, ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 2264 - 68
KARLIN, ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 77
KLYUSHNICHENKO V E ET AL: "Recombinant human insulin", JOURNAL OF CHROMATOGRAPHY, ELSEVIER SCIENCE PUBLISHERS B.V, NL, vol. 661, no. 1-2, 11 February 1994 (1994-02-11), pages 83 - 92, XP026660887, ISSN: 0021-9673, [retrieved on 19940211] *
SON ET AL.: "Effects of citraconylation on enzymatic modification of human proinsulin using trypsin and carboxypeptidase B.", BIOTECHNOL PROG., vol. 25, no. 4, July 2009 (2009-07-01), pages 1064 - 70, XP002660114, DOI: doi:10.1021/BP.195
THIM ET AL., P.N.A.S., USA, vol. 83, 1986, pages 6766 - 6770

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014084921A1 (fr) * 2012-11-27 2014-06-05 Rose Stuart Procédé de fabrication pharmaceutique et compositions
WO2014096985A3 (fr) * 2012-12-19 2015-07-16 Wockhardt Limited Composition aqueuse stable comprenant de l'insuline humaine ou un analogue ou un dérivé de celle-ci
AU2013365945B2 (en) * 2012-12-19 2017-03-02 Wockhardt Limited A stable aqueous composition comprising human insulin or an analogue or derivative thereof
CN113226284A (zh) * 2018-09-25 2021-08-06 美药星制药股份有限公司 高度纯化的重组人胰岛素(rhi)api及其生产方法

Similar Documents

Publication Publication Date Title
US7790677B2 (en) Insulin production methods and pro-insulin constructs
JP6077027B2 (ja) 精製生物活性異種タンパク質を得る方法
EP2307441B1 (fr) Procédé pour la fabrication de composés de l&#39;insuline
WO2012115638A1 (fr) Compositions de pro-insuline glargine et procédés de production d&#39;analogues de l&#39;insuline glargine à partir de celles-ci
KR102345011B1 (ko) GroES 융합을 이용한 글루카곤 유사 펩타이드-1 또는 이의 유사체의 생산방법
WO2012115640A1 (fr) Compositions liquides contenant de l&#39;insuline et leurs procédés de fabrication
KR101527528B1 (ko) 가용성 재조합 단백질의 생산, 추출 및 정제 방법
US20160030520A1 (en) Liquid insulin compositions and methods of making the same
US20120214965A1 (en) Glargine proinsulin and methods of producing glargine insulin analogs therefrom
US10100098B2 (en) Insulin production methods and proinsulin constructs
US20190352365A1 (en) Expression construct and method for producing proteins of interest
CN111718417B (zh) 含有荧光蛋白片段的融合蛋白及其用途
JP2021511785A (ja) 組換えポリペプチド生産用n末端融合パートナーおよびこれを用いた組換えポリペプチドの生産方法
CN114380903B (zh) 一种胰岛素或其类似物前体
US20160024168A1 (en) Aspart proinsulin compositions and methods of producing aspart insulin analogs
EP3344651B1 (fr) Procédé d&#39;obtention d&#39;insuline avec liaisons disulfure correctement formées
KR102064810B1 (ko) 재조합 폴리펩타이드 생산용 n-말단 융합 파트너 및 이를 이용하여 재조합 폴리펩타이드를 생산하는 방법
US20160039899A1 (en) Lis-pro proinsulin compositions and methods of producing lis-pro insulin analogs therefrom
KR102345012B1 (ko) GroES 융합을 이용한 인간부갑상선호르몬 1-34의 생산방법
CN102558356A (zh) 一种人胰岛素原融合蛋白及人胰岛素的制备方法
KR100407792B1 (ko) 인간 글루카곤 유사펩타이드를 융합파트너로 이용한재조합 단백질의 제조방법
KR102345013B1 (ko) GroES 융합을 이용한 글루카곤 유사 펩타이드-2 또는 이의 유사체의 생산방법
WO2012115637A1 (fr) Compositions de pro-insuline aspart et procédés de production d&#39;analogues de l&#39;insuline aspart
WO2012115641A1 (fr) Compositions de pro-insuline lis-pro et procédés de production d&#39;analogues d&#39;insuline lis-pro à partir de celles-ci

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: 11714871

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11714871

Country of ref document: EP

Kind code of ref document: A1