CN118063588A - Recombinant fusion protein and application thereof - Google Patents

Recombinant fusion protein and application thereof Download PDF

Info

Publication number
CN118063588A
CN118063588A CN202410363959.5A CN202410363959A CN118063588A CN 118063588 A CN118063588 A CN 118063588A CN 202410363959 A CN202410363959 A CN 202410363959A CN 118063588 A CN118063588 A CN 118063588A
Authority
CN
China
Prior art keywords
fusion protein
seq
igg
amino acid
truncated
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202410363959.5A
Other languages
Chinese (zh)
Inventor
路力生
霍永庭
闫加庆
李艳敏
罗甜
芦迪
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangdong Fapon Biopharma Inc
Original Assignee
Guangdong Fapon Biopharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Guangdong Fapon Biopharma Inc filed Critical Guangdong Fapon Biopharma Inc
Publication of CN118063588A publication Critical patent/CN118063588A/en
Pending legal-status Critical Current

Links

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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5428IL-10
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2066IL-10
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention relates to the field of biological medicine, in particular to truncated IL-10, recombinant fusion protein and application thereof. The invention provides a truncated IL-10 and recombinant IL-10-Fc fusion protein thereof, wherein the truncated IL-10 and the fusion protein thereof can effectively reduce heterogeneity, reduce immunogenicity caused by glycosylation, improve safety and prolong plasma half-life of IL-10; the fusion protein and related nucleic acid, vector, cell or pharmaceutical composition thereof can be widely used for preparing medicines for preventing and/or treating tumors.

Description

Recombinant fusion protein and application thereof
The application relates to a split application of Chinese application patent application 202111483187.1 (application number: 202111483187.1, application date: 2021-12-07, application name: a recombinant fusion protein and application thereof); meanwhile, the application claims priority of Chinese patent application 202011421111.1 (application number: 202011421111.1, application date: 2020-12-08, title: a recombinant fusion protein and application thereof) filed on 12/08 of 2020, the entire contents of which are incorporated herein by reference.
Technical Field
The invention belongs to the technical field of biological medicine. More particularly, it relates to a truncated IL-10, recombinant fusion proteins and uses thereof.
Background
Human Interleukin-10 (Intereukin-10, IL-10) was cloned and expressed by Vieira in 1991 from tetanus toxicity specific T cells, its gene was located in chromosome 1q32 region, the full length of DNA was 4.7kb, it contained 5 exons, its functional protein was a homodimer non-covalently bound by disulfide bonds, molecular weight was 35kD, each subunit contained 178 amino acids, including a 18 amino acid signal peptide, and murine IL-10 homology was about 75%. The human IL-10 receptor is a heterodimer of IL-10R1 and IL-10R2, belonging to class II cytokine receptors. IL-10 and its receptor interactions are first binding with high affinity to IL-10R1 and then through steric claimation alter binding to IL-10R2, thus mediating signal transduction of the JAK-STAT pathway.
Almost all lymphocytes are capable of synthesizing IL-10, and the main sources are secreted by mononuclear macrophages and T helper cells, and have immunosuppressive or immunostimulatory bidirectional regulation functions in various types of immune cells: on the one hand, the inhibition of Th1 cell response and cytokine synthesis, the inhibition of IL-4 and IL-5 production by Th2 cells, the inhibition of antigen presenting function of macrophages and the release of immune medium, but the inhibition of IL-17 production by Th17 cells, and the lack of direct inhibition of CD8+ T cells, are not shown; on the other hand, the proliferation and differentiation of B cells and the generation of antibodies are promoted, and the cytotoxic activity of NK cells, mast cells and T cell activity are promoted.
At present, drugs targeting IL-10 are not yet approved for the market, and most of the drugs in the research stage are used for the treatment of autoimmune diseases and chronic inflammation, whereas products used for the treatment of tumors are only Pegylated (PEG) -IL-10, such as Pegilodecakin and IL-10Synthorin. However, natural IL-10 has a half-life of only a few hours, and even after the surface is pegylated, it has a half-life of only a few days, and high doses can produce toxic side effects. Therefore, the development of the antitumor drug which takes IL-10 as a target point has significant antitumor activity and no toxic or side effect.
Disclosure of Invention
The present invention aims to solve at least to some extent one of the following technical problems:
(1) IL-10 fusion proteins are susceptible to degradation or truncation during production, resulting in heterogeneity;
(2) The presence of free cysteines in the hinge region of the IgG Fc fragment of an IL-10 fusion protein tends to cause product aggregation;
(3) Part of the N-glycosylation site may cause immunogenicity, resulting in reduced safety of the product;
(4) Human IL-10 has a short plasma half-life as a drug to treat related diseases.
The invention aims at providing a truncated IL-10, wherein the truncated IL-10 lacks Ser and Pro at the N terminal compared with wild type IL-10, and the amino acid sequence of the wild type IL-10 is shown as SEQ ID NO:5, the amino acid sequence of the truncated IL-10 is shown as SEQ ID NO: shown at 9.
The invention also provides a recombinant fusion protein comprising IL-10, a connecting peptide and an IgG Fc fragment, wherein the C-terminal end of the IL-10 is connected to the N-terminal end of the IgG Fc fragment through the connecting peptide, and the IL-10 lacks Ser and Pro at the N-terminal end compared with wild type IL-10.
In some embodiments, the sequence of the connecting peptide is as set forth in SEQ ID NO:6 or SEQ ID NO: shown at 10.
In some embodiments, the hinge region of the IgG Fc fragment is substituted with Ser at Cys at position 220 according to EU numbering.
In some embodiments, the IgG Fc fragment is substituted with Ala at Leu at position 235, gly at position 237, or Ala at position 327 according to EU numbering.
The invention also provides nucleic acids, vectors, cells or pharmaceutical compositions related to the fusion proteins.
The invention also relates to the application of the fusion protein, the nucleic acid, the vector, the cell or the pharmaceutical composition in preparing medicines for preventing and/or treating tumors.
Drawings
FIG. 1 is a schematic diagram of the constructed pCDNA3.4A-R0356 vector containing the target gene.
FIG. 2 is a schematic diagram of the constructed pCDNA3.1-M1 vector containing the target gene.
FIG. 3 is a schematic diagram of the constructed pCDNA3.1-M2 vector containing the target gene.
FIG. 4 is a schematic diagram of the constructed pCDNA3.1-M3 vector containing the target gene.
Fig. 5 is a graph of SEC detection results for R0356.
Fig. 6 is a graph of SEC detection results for M1.
Fig. 7 is a graph of SEC detection results for M2.
Fig. 8 is a graph of SEC detection results for M3.
Fig. 9 is a graph of the mass spectrum detection results of R0356.
FIG. 10 is a graph showing the results of a measurement of binding of recombinant IL-10-Fc fusion protein to IL-10 receptor IL-10R1 at the cellular level; in the figure, "Isotype" represents an IgG1 protein that does not fuse IL-10; "HillSlope" represents the slope of the curve at the EC50 and "Span" represents the upper plateau value of the curve at the EC 50.
FIG. 11 is a graph showing the results of reporter gene detection; "HillSlope" represents the slope at the EC 50.
Fig. 12 is a graph of tumor growth.
Detailed Description
The present invention is further illustrated below with reference to specific examples, which are not intended to limit the invention in any way. Unless specifically stated otherwise, the reagents, methods and apparatus employed in the present invention are those conventional in the art.
Reagents and materials used in the following examples are commercially available unless otherwise specified.
The invention relates to a recombinant fusion protein, comprising IL-10, a connecting peptide and an IgG Fc fragment, wherein the C-terminal end of the IL-10 is connected to the N-terminal end of the IgG Fc fragment through the connecting peptide, and the IL-10 lacks Ser and Pro at the N-terminal end compared with wild type IL-10.
The present invention reduces or eliminates product heterogeneity due to degradation or truncation of IL-10 fusion proteins by truncating both Ser and Pro amino acids at the N-terminus of IL-10.
In some embodiments, the Fc is selected from any one of IgG1 Fc, igG2 Fc, igG3Fc, or IgG4 Fc.
In a preferred embodiment, the Fc is an IgG1 Fc.
In the present invention, igG Fc comprises IgG CH2 and IgG CH3 domains. IgG1 Fc, igG2 Fc, igG3Fc, igG4 Fc represent the Fc of IgG1, igG2, igG3 and IgG4, respectively.
In the present invention, the term "Fc" is used to define a C-terminal region of an antibody heavy chain that contains at least a portion of a constant region.
In some embodiments, the sequence of the connecting peptide is as set forth in SEQ ID NO:6 or SEQ ID NO: shown at 10.
The invention removes the generated unexpected N-glycosylation site by modifying the connecting peptide, reduces immunogenicity caused by glycosylation and improves safety.
In some embodiments, the hinge region of the IgG Fc fragment is substituted with Ser at Cys at position 220 according to EU numbering.
In some embodiments, the IgG Fc fragment is substituted with Ala at Leu at position 235, gly at position 237, or Ala at position 327 according to EU numbering. Thereby reducing product aggregation.
Unless otherwise specified herein, numbering of amino acid residues of an IgG Fc fragment is according to the EU numbering system, also known as the EU index, as set forth in Kabat et al.,Sequences of Proteins of ImmunologicalInterest,5th Ed.Public Health Service,National Institutes of Health,Bethesda,MD,1991.
In some embodiments, the fusion protein has an amino acid sequence as set forth in SEQ ID NO: 2. SEQ ID NO:3 or SEQ ID NO: 4.
In the invention, the amino acid sequence of R0356 is shown as SEQ ID NO:1 is shown in the specification; in R0356, the amino acid sequence of IL-10 (wild type IL-10) is shown in SEQ ID NO:5 is shown in the figure; the amino acid sequence of the connecting peptide is shown in SEQ ID NO:6 is shown in the figure; the amino acid sequence of the IgG Fc fragment is shown in SEQ ID NO: shown at 7.
In some embodiments, the fusion protein is M1, M2, or M3.
In some embodiments, the amino acid sequence of M1 is as set forth in SEQ ID NO:2 is shown in the figure; in M1, the amino acid sequence of IL-10 is shown in SEQ ID NO: shown as 9; the amino acid sequence of the connecting peptide is shown in SEQ ID NO:6 is shown in the figure; the amino acid sequence of the IgG Fc fragment is shown in SEQ ID NO: shown at 7.
In some embodiments, the amino acid sequence of M2 is as set forth in SEQ ID NO:3 is shown in the figure; in M2, the amino acid sequence of IL-10 is shown in SEQ ID NO: shown as 9; the amino acid sequence of the connecting peptide is shown in SEQ ID NO:6 is shown in the figure; the amino acid sequence of the IgG Fc fragment is shown in SEQ ID NO: shown at 8.
In some embodiments, the amino acid sequence of M3 is as set forth in SEQ ID NO:4 is shown in the figure; in M3, the amino acid sequence of IL-10 is shown in SEQ ID NO: shown as 9; the amino acid sequence of the connecting peptide is shown in SEQ ID NO:10 is shown in the figure; the amino acid sequence of the IgG Fc fragment is shown in SEQ ID NO: shown at 8.
In the present invention, "wild-type IL-10" refers to naturally occurring IL-10, which is a homodimer made up of two α -helical monomer domains, as opposed to "modified IL-10", which is a naturally occurring IL-10 that has been modified, e.g., to alter one or more of its properties, such as stability, affinity. The modified IL-10 molecule may, for example, comprise modifications in the amino acid sequence, such as amino acid substitutions, deletions or insertions.
In the present invention, a "linker peptide" comprises one or more amino acids, typically peptides of about 2-20 amino acids, as known in the art or as described herein. Suitable non-immunogenic connecting peptides include, for example, (GS) n linker, where n is an integer from 1 to 10.
In the present invention, an amino acid "substitution" refers to the replacement of one amino acid in a polypeptide with another amino acid. In one embodiment, the amino acid is replaced with another amino acid having similar structural and/or chemical properties, e.g., a conservative amino acid substitution. "conservative" amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. For example, nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; while negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Non-conservative substitutions may require exchanging members of one of these classes for members of the other class. For example, amino acid substitutions may also result in the replacement of one amino acid with another amino acid having a different structural and/or chemical property, e.g., the replacement of an amino acid from one group (e.g., polar) with another amino acid from a different group (e.g., basic). Genetic or chemical methods well known in the art may be used to generate amino acid substitutions.
In the present invention, the term "modification" refers to any manipulation of the peptide backbone (e.g., amino acid sequence) or post-translational modification (e.g., glycosylation) of the polypeptide. Modifications also include substitutions, deletions or insertions of amino acids in the amino acid sequence.
The invention also relates to a nucleic acid encoding the fusion protein.
The invention also relates to a vector carrying the nucleic acid.
In the present invention, the term "vector" is a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes vectors that are self-replicating nucleic acid structures and vectors that integrate into the genome of a host cell into which they are introduced.
The invention also relates to a cell containing said nucleic acid or said vector.
The invention also relates to a pharmaceutical composition comprising said fusion protein, said nucleic acid, said vector or said cell.
In a preferred embodiment, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier.
In the present invention, the term "pharmaceutical composition" is in a form that allows the biological activity of the active ingredient to be effective and does not comprise additional ingredients that have unacceptable toxicity to the subject to which the composition is to be administered.
In the present invention, the term "pharmaceutically acceptable carrier" may include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible for extending the shelf life or efficacy of the antibodies.
The invention also relates to the application of the fusion protein, the nucleic acid, the vector, the cell or the pharmaceutical composition in preparing medicines for preventing and/or treating tumors.
In the present invention, the term "treatment" refers to the administration of an effective amount of a therapeutically active substance with the aim of alleviating, ameliorating, inhibiting or eradicating (curing) symptoms or disease states. For the prevention or treatment of a disease, the appropriate dosage of the fusion protein of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the route of administration, the weight of the patient, the type of fusion protein, the severity and course of the disease, whether the fusion protein is administered for prophylactic or therapeutic purposes, previous or concurrent therapeutic interventions, the patient's clinical history and response to the fusion protein, and the discretion of the attending physician. The practitioner responsible for administration will determine the concentration of the active ingredient in the composition and the appropriate dosage for the individual subject at any event. Various dosing regimens are contemplated herein, including, but not limited to, single or multiple administrations at various points in time, bolus administrations, and pulse infusion.
The invention has the following beneficial effects:
The invention constructs a gene recombinant fusion protein IL-10-Fc of IL-10 and Fc fragment of an IgG antibody, and reduces product heterogeneity by modifying IL-10, such as truncating; furthermore, the non-beneficial N-glycosylation site is removed by modifying the connecting peptide, so that the immunogenicity caused by glycosylation is reduced, furthermore, the Fc fragment is subjected to site-directed mutagenesis, the high polymer is reduced, the ADCC effect is reduced, the safety is improved, the half-life of plasma is prolonged through Fc fusion IL-10, the administration frequency can be reduced, and the curative effect of the safety of the medicine is improved.
EXAMPLE 1 Gene construction of recombinant IL-10-Fc fusion proteins
The amino acid sequences of R0356, M1, M2, M3 and M4 in Table 1 were translated into DNA sequences, and genes were optimized and synthesized conventionally according to the codons of the Expi CHO or Expi 293F cells, after which HindIII cleavage site and 5'UTR protecting base ([ CCGC ]) were added in sequence at the 5' end; the 3' end is added with a 3' UTR stop codon ([ TGATGA ]), an EcoRI restriction site and a 3' UTR protective base ([ CGG ]), the target gene amplified by the PCR under the conventional condition is cloned to a vector pCDNA3.1A or pCDNA3.4A through 5' EcoRI and 3' HindIII, and the schematic diagram of the vector (pCDNA3.4A-R0356, pCDNA3.1-M1, pCDNA3.1-M2 and pCDNA3.1-M3) containing the target gene is constructed as shown in the figures 1-4.
The DNA molecule of interest and the vector molecule were subjected to HindIII (NEB) and EcoRI (NEB) double cleavage and were transduced into DH 5. Alpha. Competent bacteria (NEB) after ligation with T4 DNA ligase (TAKARA). And (3) culturing by using an LB culture solution of ampicillin, selecting clones for sequencing, selecting thalli with correct sequencing for seed preservation, and performing expansion culture of the thalli, wherein the expanded thalli are used for extracting plasmids.
TABLE 1 amino acid sequences of R0356, M1, M2, M3 and M4
Fusion proteins IL-10 Connecting peptide IgG Fc fragment
R0356(SEQ ID NO:1) SEQ ID NO:5 SEQ ID NO:6 SEQ ID NO:7
M1(SEQ ID NO:2) SEQ ID NO:9 SEQ ID NO:6 SEQ ID NO:7
M2(SEQ ID NO:3) SEQ ID NO:9 SEQ ID NO:6 SEQ ID NO:8
M3(SEQ ID NO:4) SEQ ID NO:9 SEQ ID NO:10 SEQ ID NO:8
M4(SEQ ID NO:11) SEQ ID NO:5 SEQ ID NO:6 SEQ ID NO:8
Wherein, the structures of R0356, M1, M2 and M3 are IL-10-connecting peptide-IgG Fc; m4 has the structure of IgG Fc-connecting peptide-IL-10.
EXAMPLE 2 expression and purification of recombinant IL-10-Fc fusion proteins
1. Experimental method
In this example, the plasmid extracted in example 1 is used, and the Expi293F cell is taken as an expression system for describing the cell transfection and transient expression technical scheme, and other conventional technical processes should be regarded as the protection scope of the present invention.
Selecting excellent-state and activity greater than 95% of Expi293F (Thermo) cells, adjusting the density to 3×10 6 cells/mL by using Dynamis culture medium (GIBCO), gently shaking and sub-packaging the cells (transfection system 90%), wherein the cell volume in a shake flask is not more than 1/3 of the shake flask specification, and placing into a shaking table for standby. Calculating the volume of the transfection buffer solution opti-MEM according to the volume of transfected cells, wherein the volume is 1/10 of that of a transfection system; calculating the amount of the transfection reagent PEI, wherein the proportion of the amount of the transfection reagent PEI is 3 mu L/mL of transfected cells; the total amount of transfected DNA was calculated at a ratio of 1. Mu.g/mL transfected cells. The specific transfection procedure was as follows:
Taking 150 mL centrifuge tube, adding transfection buffer opti-MEM with the concentration of 10% of the transfection system, adding plasmid, mixing uniformly, filtering, standing for 5min, adding PEI into the DNA suspension, mixing gently (mixing gently for 2-3 times), and standing for 15-20min. And then the compound is gently added into the packaged cells, and the shake flask is gently shaken while the compound is added, so that the cell transfection is completed. After the transfected cells were placed in a shaker at 37℃and 8% CO 2 at 120rpm for 24 hours, the culture supernatant was collected on day 15 by adding the feed medium to the volume of 5% transfected cells.
Then, the culture supernatant was subjected to affinity chromatography purification, and the affinity chromatography steps were as follows:
Chromatography column 1 (GE HEALTHCARE, XK/20, column Volume (CV), 53 ml) loaded with chromatography packing 1 (GE HEALTHCARE, MABSELECT SuRe LX) was regenerated in a chromatography system (GE HEALTHCARE, AKTA pure 150), equilibrated with 20mM PBS for 2CV, and the UV detector (UV Monitor) was reset to load the fermentation supernatant of the Expi293 cells in a bubble sensitive manner. After the completion of the loading, the column was washed with 20mM PBS for 3CV, then eluted with 100% buffer 20mM NaAc, pH3.4 step for 3CV, the 40 mAu-40 mAu 280nm UV absorbing fraction was collected, and 1.8% neutralization buffer was previously added to the collection tube to bring the final pH to the range of 6.0-7.0, then washed in place for 4CV for 5min, then washed with buffer A flowing downward for 5CV, and then stored with 20% ethanol buffer.
The method adopts a chromatographic column preloaded with PG200 filler to carry out molecular sieve action on the sample eluted by the chromatographic column, and comprises the following steps:
And (3) further performing molecular sieve purification on the captured elution sample, and performing sterilization and filtration on the affinity elution sample by using a 0.22um filter membrane for later use. The chromatographic column 2 (Boguron, BXK/100, column Volume (CV), 1825 ml) containing the chromatographic packing 2 (Boguron, PG 200) was equilibrated with 20mM PBS for 2CV in a chromatographic system (GE HEALTHCARE, AKTA AVANT 150), the UV detector (UV Monitor) was reset, the sample after R0673 affinity elution was loaded in a bubble-sensitive manner, the column 2CV was washed with 20mM PBS after the loading was completed, the 20 mAu-20 mAu 280nm UV absorbing fraction was collected, and the molecular integrity was analyzed by mass spectrometry. Samples were harvested after 15 days of incubation and tested for purity and intact molecular weight by Size Exclusion Chromatography (SEC) and Mass Spectrometry (MS).
The method for detecting purity by using the size exclusion chromatography comprises the following steps: the standard was diluted to 1.0mg/mL with mobile phase A (50 mM phosphate+300 mM potassium chloride, pH 7.0.+ -. 0.1), and the mixture was mixed and then placed in a liner tube. The instrument setting parameters are shown in the following table, and the mobile phase A-reference sample-to-be-detected sample-reference sample sequential sample injection detection is performed. The results were quantitatively analyzed by peak area normalization and the peak area percentages of monomer, polymer, low molecular weight hybrid were calculated. The area percentage of the monomer peak is taken as the sample purity, the high polymer and the area percentage of the low molecular weight impurity peak as the content of the sample impurity.
2. Experimental results
The SEC detection results of R0356, M1, M2 and M3 are shown in fig. 5-8 and table 2 in sequence, which shows that the high polymers of M2 and M3 are significantly reduced compared with R0356 and M1, indicating that M2 and M3 effectively reduce the generation of polymers.
Table 2 SEC high polymer detection results for M1, M2 and M3
Sample name High polymer (%)
R0356 14.73
M1 14.44
M2 6.97
M3 9.51
The mass spectrum of R0356 is shown in FIG. 9, which shows that about 50% of the molecules in R0356 were truncated; the mass spectrum detection results of M1, M2 and M3 are shown in Table 3, and the results show that M1, M2 and M3 are kept in an intact state, no degradation or truncation is observed, and the heterogeneity is effectively reduced.
Table 3 mass spectrometric detection results for M1, M2 and M3
Sample name Description of the invention Theoretical molecular weight Measured molecular weight Difference (Da) Difference (ppm)
M1 Half antibody molecules 45009.56 45013.34 3.78 83.98
M2 Half antibody molecules 44993.50 44997.21 3.71 82.46
M3 Half antibody molecules 45107.60 45110.81 3.21 71.16
EXAMPLE 3 detection of glycosylation sites
1. Experimental method
The detection amino acid sequence is SEQ ID NO:1-4, and detecting glycosylation by using NetNGlyc 1.0Server module in ExPASy.
2. Experimental results
The glycosylation site detection results show that R0356, M1 and M2 have an abnormal glycosylation site NGSG formed by the tail end of IL-10 and Linker sequence in addition to the 2 glycosylation sites naturally occurring in IL-10 and IgG Fc; in contrast, M3 has only 2 glycosylation sites naturally occurring in IL-10 and IgG Fc, and no glycosylation site NGSG is detected, so that immunogenicity due to glycosylation is reduced, and safety is improved.
EXAMPLE 4 determination of binding of recombinant IL-10-Fc fusion protein to IL-10 receptor IL-10R1 at the cellular level
1. Experimental method
In this example, recombinant IL-10-Fc fusion proteins produced and purified in examples 1 and 2 were selected and tested for binding of recombinant IL-10-Fc fusion proteins to IL-10 receptor IL-10R1 at the cellular level based on cells overexpressing IL-10R 1.
2X 10 5 cells/well of CHO-IL10R cells were plated on a VU bottom 96 well plate overnight; washing the plate with 200. Mu.L PBS, centrifuging, removing supernatant, adding 200. Mu.L precooled 3% BSA-PBS blocking solution, and blocking at 2-8deg.C for 1 hr; after washing the plates with the waste liquid, samples of 3-fold serial dilutions of R0356 (labeled R0356 (plate 1) and R0356 (plate 2) in FIG. 10), M1 (M1-E1 and M1-E2 in FIG. 10), M2 (M2-E1 and M2-E2 in FIG. 10) M3, M4, isotype (IgG 1 protein without IL-10 fusion, i.e.homotype IgG1 control) and blank (without any protein addition, i.e.blank) were added sequentially from 15. Mu.g/mL, containing a total of 11 dilution points at zero, 100. Mu.l/well, multiple wells, and incubated at 2-8deg.C for 2h; after washing the plate with the discard solution, 1:15000 dilution Goat Anti-Human IgG Fc (HRP) secondary antibody, 100 μl/well, incubation at 2-8deg.C for 1h; after washing the plate with waste liquid, add the following 1:15000 dilution of Goat Anti-Human IgG Fc (HRP) secondary antibody, 100 μl/well, incubation at 2-8deg.C for 1h; after washing the plate with the waste liquid, 100. Mu.l/well of TMB was added to each well, and the color development was stopped with 100. Mu.l/well of stop solution (1M H 2SO4) at 37℃for 10-20 min in the absence of light. Absorbance at a450nm was collected with a microplate reader and EC50 was calculated using GRAPHPAD PRISM 5.0.0 software.
2. Experimental results
The results of the measurement of binding of recombinant IL-10-Fc fusion protein to IL-10 receptor IL-10R1 at the cellular level are shown in FIG. 10, which shows that the EC50 of R0356, M1, M2 and M3 are comparable.
Example 5 reporter Gene detection of biological Activity
1. Experimental method
In this example, recombinant IL-10-Fc fusion proteins produced and purified in examples 1 and 2 were selected, and a reporter gene system H_IL10 receptor 293cell line (Genomeditech) in which IL-10R and STAT3 signal pathways were connected in series was used as a detection system, which stimulated an increase in the expression of cellular luciferase when IL-10-containing samples were bound to IL-10R on the surface of the system. The specific detection steps are as follows:
Firstly, 50 mu l/well of H_IL10 receptor 293cell line cells of 2X 10 5 cells/well are paved into 96-well plates; immediately after the dilution of R0356, M1 (M1-E2 in FIG. 11), M2 (M2-E2 in FIG. 11), M3 (M3-E2 in FIG. 11) and M4 (M4-E2 in FIG. 11) with the assay buffer in sequence 2-fold from 10. Mu.g/mL, a total of 10 dilution points at zero point, 50. Mu.l/well were added, mixed and incubated in a CO 2 incubator at 37℃for 16h; mu.l of luciferase reporter detection reagent (Cat#RG051M, beyotime) was added to each well and incubated for 10min, fluorescence intensity values were collected with a microplate reader and EC50 was calculated using GRAPHPAD PRISM 5.0.0 software.
2. Experimental results
The results of the reporter gene detection are shown in FIG. 11, and the results show that the EC50 of R0356, M1, M2 and M3 are equivalent, the biological activity of M3 is better than that of R0356, and the relative biological activity is 106.5%.
EXAMPLE 6 in vivo evaluation of recombinant IL-10-Fc fusion proteins for anti-tumor
1. Experimental method
Taking mouse colon cancer CT26 cells in logarithmic growth phase, and regulating the cell concentration to be 1X 10 6/mL. Female BALB/C mice were inoculated subcutaneously with CT26 cells at a volume of 0.1 mL/mouse. Inoculation mice were randomized into 3 groups of 8 animals per group by tumor volume on day 0 (D0) and day 9, and dosing was initiated (dosing schedule is shown in table 4). Wherein M1, M2 and M3 are M1, M2 and M3, respectively, in this example, R0511 is isotype IgG1 control.
Table 4 dosing regimen
Tumor volumes were measured and recorded starting on the day of dosing, after which tumor long and short diameters were measured 2 times per week with vernier calipers. Tumor volume tumor growth inhibition TGI (%) = [1- (Ti-T0)/(Vi-V0) ]x100 was calculated by the formula, ti was the average tumor volume of the treatment group on day i, T0 was the average tumor volume of the treatment group at the start of treatment, vi was the average tumor volume of the solvent control group on day i, and V0 was the average tumor volume of the solvent control group at the start of treatment. When each mouse reached the end of the experiment (tumor volume exceeded 2000mm 3 reached the end of the kernel-day), the mice were sacrificed by cervical dislocation and survival curves were recorded.
2. Experimental results
The average tumor volumes of administration of M1, M2, M3, R0511 and R0356 are shown in table 5, and the results show that the average tumor volume of M3 is significantly smaller than R0356, indicating that M3 has an effect of inhibiting tumor cell growth.
Table 5 average tumor volume results for administration of M1, M2, M3, R0511 and R0356
The tumor growth curves are shown in fig. 12, and the results show that the antitumor effect of M3 is stronger than that of R0356, the antitumor effects of M2 and R0356 are equivalent, and the antitumor effect of M1 is the weakest.
The above examples are preferred embodiments of the present invention, but the embodiments of the present invention are not limited to the above examples, and any other changes, modifications, substitutions, combinations, and simplifications that do not depart from the spirit and principle of the present invention should be made in the equivalent manner, and the embodiments are included in the protection scope of the present invention.

Claims (10)

1. A truncated IL-10, wherein said truncated IL-10 lacks N-terminal Ser and Pro as compared to wild-type IL-10, and wherein said wild-type IL-10 has an amino acid sequence as set forth in SEQ ID NO:5, the amino acid sequence of the truncated IL-10 is shown as SEQ ID NO: shown at 9.
2. A recombinant fusion protein consisting of IL-10, a linker peptide and an IgG Fc fragment, wherein the C-terminus of IL-10 is linked to the N-terminus of the IgG Fc fragment by the linker peptide, and wherein the IL-10 lacks Ser and Pro at the N-terminus compared to wild-type IL-10.
3. A recombinant fusion protein consisting of IL-10, a linker peptide and an IgG Fc fragment, wherein the C-terminus of IL-10 is linked to the N-terminus of the IgG Fc fragment by the linker peptide.
4. A fusion protein according to claim 2 or 3, wherein the sequence of the linker peptide is as set forth in SEQ ID NO:6 or SEQ ID NO: shown at 10.
5. The fusion protein of any one of claims 2-4, wherein the hinge region of the IgG Fc fragment is substituted with Ser at Cys at position 220 according to EU numbering.
6. The fusion protein of any one of claims 2-5, wherein the IgG Fc fragment is EU-numbered with Leu at position 235 substituted with Ala, gly at position 237 substituted with Ala, and Ala at position 327 substituted with gin.
7. The fusion protein of any one of claims 2-6, wherein the fusion protein has an amino acid sequence as set forth in SEQ ID NO: 2. SEQ ID NO:3 or SEQ ID NO: 4.
8. A biomaterial, characterized in that the biomaterial is selected from any one of (i) to (iii):
A nucleic acid encoding the truncated IL-10 of claim 1 or the fusion protein of any one of claims 2-7;
(ii) a vector carrying the nucleic acid of (i) above;
(iii) a cell carrying the nucleic acid of (i) or containing the vector of (ii).
9. A pharmaceutical composition comprising a truncated IL-10 according to claim 1 or a fusion protein according to any one of claims 2-7, a biomaterial according to claim 8.
10. Use of a truncated IL-10 according to claim 1 or a fusion protein according to any one of claims 2-7, a biomaterial according to claim 8 or a pharmaceutical composition according to claim 9 for the manufacture of a medicament for the prevention and/or treatment of tumors.
CN202410363959.5A 2020-12-08 2021-12-07 Recombinant fusion protein and application thereof Pending CN118063588A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202011421111 2020-12-08
CN2020114211111 2020-12-08
CN202111483187.1A CN114621353B (en) 2020-12-08 2021-12-07 Recombinant fusion protein and application thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN202111483187.1A Division CN114621353B (en) 2020-12-08 2021-12-07 Recombinant fusion protein and application thereof

Publications (1)

Publication Number Publication Date
CN118063588A true CN118063588A (en) 2024-05-24

Family

ID=81897990

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202111483187.1A Active CN114621353B (en) 2020-12-08 2021-12-07 Recombinant fusion protein and application thereof
CN202410363959.5A Pending CN118063588A (en) 2020-12-08 2021-12-07 Recombinant fusion protein and application thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202111483187.1A Active CN114621353B (en) 2020-12-08 2021-12-07 Recombinant fusion protein and application thereof

Country Status (1)

Country Link
CN (2) CN114621353B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116125074A (en) * 2022-12-13 2023-05-16 北京东方百泰生物科技股份有限公司 Method for detecting binding activity of IL-10-Fc fusion protein and receptor thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101948543B (en) * 2010-08-30 2013-01-16 中国科学技术大学 Fusion protein and coding gene and application thereof
CN101962413B (en) * 2010-09-21 2013-03-13 中国科学技术大学 Fusion protein with transdermal capability and interleukin-10 activity as well as coding gene and application thereof
CN108948207B (en) * 2017-05-22 2020-11-13 杭州博虎生物科技有限公司 Human interleukin 10-Fc fusion protein and coding gene and application thereof
WO2020097946A1 (en) * 2018-11-18 2020-05-22 杭州博虎生物科技有限公司 Recombinant human interleukin-10 fusion protein and application thereof

Also Published As

Publication number Publication date
CN114621353B (en) 2024-04-02
CN114621353A (en) 2022-06-14

Similar Documents

Publication Publication Date Title
KR102609197B1 (en) Interleukin 15 protein complex and use thereof
Han et al. IL-15: IL-15 receptor alpha superagonist complex: high-level co-expression in recombinant mammalian cells, purification and characterization
JP6923115B2 (en) Highly glycosylated human blood coagulation factor VIII fusion protein, its preparation method, and its use
AU2018247255A1 (en) Single chain fc fusion proteins
CN110128525B (en) FGF21 variants, fusion proteins, and uses thereof
CN113038964A (en) Ultra-long-acting insulin-FC fusion protein and use method thereof
DK2633865T3 (en) USE OF INTERLEUKIN-22 IN THE TREATMENT OF VIRAL HEPATITIS
US20240131114A1 (en) Nerve growth factor fusion protein, preparation method and use thereof
CA2743394C (en) Il-4-derived peptides for modulation of the chronic inflammatory response and treatment of autoimmune diseases
CN110256575B (en) Long-acting recombinant human growth hormone fusion protein and engineering cell thereof
EP0567599A1 (en) Megakaryocyte stimulating factors
CN113105562B (en) Application of mutant single-chain human coagulation factor VIII in preparation of fusion protein
US10562954B2 (en) Fusion protein inhibiting TACI-BAFF complex formation and preparation method therefor and use thereof
CN114621353B (en) Recombinant fusion protein and application thereof
CN113613675B (en) TACI-Fc fusion protein pharmaceutical formulations
CN106008717B (en) Long-acting recombinant GLP-1 fusion protein and preparation method and application thereof
CA2735085C (en) Fc gamma iib receptor for the treatment of b cell mediated multiple sclerosis
CN114751962B (en) Staple peptide, preparation method and pharmaceutical application thereof
WO2021063313A1 (en) Fc variant having altered effector function and fusion protein thereof
TW202216747A (en) Fusion protein comprising glucagon-like peptide-1 and interleukin-1 receptor antagonist, and use thereof
EP2578603A1 (en) B cell activating factor antagonist and preparation method and use thereof
WO2006138610A2 (en) Interferon-alpha/beta binding fusion proteins and therapeutic uses thereof
WO2023025123A1 (en) Gdf15 fusion protein and use thereof
WO2023125881A1 (en) Fusion protein of glp-1 and gdf15 and use thereof
WO2023138573A1 (en) Complex of interleukin 21 and receptor thereof

Legal Events

Date Code Title Description
PB01 Publication