WO2023134742A1 - Three-target anti-tumor drug, and preparation method therefor and use thereof - Google Patents

Three-target anti-tumor drug, and preparation method therefor and use thereof Download PDF

Info

Publication number
WO2023134742A1
WO2023134742A1 PCT/CN2023/072066 CN2023072066W WO2023134742A1 WO 2023134742 A1 WO2023134742 A1 WO 2023134742A1 CN 2023072066 W CN2023072066 W CN 2023072066W WO 2023134742 A1 WO2023134742 A1 WO 2023134742A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
domain
positions
fusion protein
cld18a2
Prior art date
Application number
PCT/CN2023/072066
Other languages
French (fr)
Chinese (zh)
Inventor
姚高峰
黄岩山
Original Assignee
浙江道尔生物科技有限公司
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 浙江道尔生物科技有限公司 filed Critical 浙江道尔生物科技有限公司
Publication of WO2023134742A1 publication Critical patent/WO2023134742A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material

Definitions

  • the invention relates to the field of biology, in particular to a three-target antitumor drug specifically targeting CLD18A2, human serum albumin and CD3, its preparation method and application.
  • Claudin 18 is a transmembrane protein with a molecular weight of about 28kD, located in the tight junction of epithelium and endothelium, and is tightly connected between adjacent cells. In normal epithelial tissue, claudin on the cell surface is difficult to access due to the tight intercellular space, while the intercellular space in tumor cells is relatively loose. Therefore, claudin on tumor cells has become a potential target for extracellular antibodies and immunotherapy.
  • CLD18 has four hydrophobic regions, which form two extracellular domains as transmembrane regions, in which hydrophobic region 1 and hydrophobic region 2 surround to form extracellular domain 1, and hydrophobic region 3 and hydrophobic region 4 surround to form extracellular domain 2.
  • the human CLD18 gene has two different exon 1, which can be alternatively spliced after transcription to generate two protein isoforms with different sequences only at the N-terminus: CLD18A1 and CLD18A2.
  • CLD18A1 is expressed in normal lung tissue, whereas CLD18A2 is expressed only in gastric tissue; more importantly, CLD18A2 is restricted to differentiated short-lived gastric epithelial cells but absent from gastric stem cells (Niimi T, et al. Biol .2001;21(21):7380-7390).
  • CLD18A2 is highly expressed in primary gastric cancer and its metastatic cancer types, and its activated expression is often observed in pancreatic cancer, esophageal cancer, ovarian cancer, and lung cancer. These properties of CLD18A2 make it a potential target for clinical treatment of gastric cancer and other related tumors.
  • the object of the present invention is to provide a three-target anti-tumor drug specifically targeting CLD18A2, human serum albumin and CD3, its preparation method and application.
  • a fusion protein comprising: an anti-CLD18A2 domain, an anti-human serum albumin (HSA) domain and an anti-CD3 domain;
  • the anti-CLD18A2 domain is a single-domain antibody with SEQ ID NO: CDR1 shown in No. 26-33 in 1, CDR2 shown in No. 51-57 in SEQ ID NO: 1, CDR3 shown in No. 96-109 in SEQ ID NO: 1;
  • Said anti The human serum albumin domain is a single domain antibody with CDR1 shown in positions 155-162 in SEQ ID NO:1, CDR2 shown in positions 180-187 in SEQ ID NO:1, and in SEQ ID NO:1 CDR3 indicated at positions 226-233.
  • the anti-CLD18A2 domain is a single domain antibody, and its framework region (FR) has a P ⁇ A mutation in the amino acid sequence; preferably, its FR1 region has a P ⁇ A mutation; more Preferably, the P ⁇ A mutation corresponds to the 14th position of SEQ ID NO:1.
  • the anti-human serum albumin domain is a single domain antibody, and its framework region (FR) has a P ⁇ A mutation in its amino acid sequence; preferably, its FR1 region has a P ⁇ A mutation Mutation; more preferably, the amino acid of its P ⁇ A mutation is the 143rd amino acid corresponding to the sequence of SEQ ID NO:1.
  • the anti-CD3 domain is a single chain antibody.
  • the anti-CD3 domain has HCDR1 shown in positions 279-286 in SEQ ID NO: 1, HCDR2 shown in positions 304-313 in SEQ ID NO: 1, SEQ ID NO: 1 HCDR3 shown in No. 352-367 in ID NO:1, LCDR1 shown in No. 419-427 in SEQ ID NO:1, LCDR2 shown in No. 445-447 in SEQ ID NO:1, SEQ ID NO : LCDR3 shown in bits 484-492 in 1.
  • the anti-CD3 domain is located at the N-terminal or C-terminal of the anti-human serum albumin domain.
  • amino acid sequence of the anti-CLD18A2 domain is shown in positions 1-120 of SEQ ID NO:1.
  • amino acid sequence of the anti-human serum albumin domain is shown in positions 130-244 of SEQ ID NO:1.
  • amino acid sequence of the anti-CD3 domain is shown in positions 254-502 of SEQ ID NO:1.
  • the amino acid sequences of the anti-CLD18A2 domain, the anti-human serum albumin domain and the anti-CD3 domain are directly connected, or connected through a linker (connecting peptide); preferably, the The amino acid sequence of the fusion protein is shown in SEQ ID NO:1.
  • the linker includes, but is not limited to: a flexible polypeptide containing glycine (G), serine (S) and/or alanine (A).
  • the two linkers when linked by two linkers, are the same; alternatively, the two linkers are different.
  • the linker is, for example, 2A, such as P2A, E2A, T2A.
  • the anti-CLD18A2 VHH, anti-HSA VHH and anti-CD3 scFv, from N-terminus to C-terminus are: CLD18A2VHH, anti-HSA VHH and anti-CD3 scFv.
  • the anti-CLD18A2 VHH, anti-HSA VHH and anti-CD3 scFv, from N-terminus to C-terminus are: CLD18A2VHH, anti-CD3 scFv and anti-HSA VHH.
  • a polynucleotide or an expression construct comprising the polynucleotide (such as expression vector), the polynucleotide encodes the fusion protein.
  • a fusion protein expression system contains the expression construct or the exogenous polynucleotide integrated in its genome.
  • the expression system is a host cell.
  • the expression system is a eukaryotic expression system.
  • the eukaryotic expression system is an animal cell expression system or a yeast expression system.
  • the animal cell expression system includes (but not limited to): CHO, NSO, BHK, HEK-293 or PER-C6.
  • the animal cell expression system is CHO.
  • the expression system is a prokaryotic expression system, such as a prokaryotic host cell.
  • a method for preparing the fusion protein comprising: using the expression system to express the fusion protein; preferably, the method further includes: isolating the fusion protein .
  • a method for modifying a CLD18A2 binding molecule to improve its therapeutic effect comprising: using an anti-CLD18A2 domain as a CLD18A2 binding molecule, and linking it to an anti-human serum albumin domain and an anti-CD3 domain ;
  • the anti-CLD18A2 structural domain is a single domain antibody, with CDR1 shown in the 26th-33rd position in SEQ ID NO: 1, CDR2 shown in the 51-57th position in SEQ ID NO: 1, SEQ ID NO: 1
  • the CDR3 shown in the 96th-109th position; the anti-human serum albumin domain is a single domain antibody, with the CDR1 shown in the 155th-162th position in SEQ ID NO:1, and the 180th position in SEQ ID NO:1 - CDR2 shown in position 187, CDR3 shown in positions 226-233 in SEQ ID NO:1.
  • a pharmaceutical composition which includes: said fusion protein.
  • a pharmaceutical composition wherein said fusion protein expression system, or its culture.
  • a kit which includes: a container or package, and the fusion protein therein.
  • a kit which includes: a container or package, and the expression system of the fusion protein or its culture located therein.
  • kits comprising: a container or package, and a The pharmaceutical composition described in.
  • a method for alleviating or treating CLD18A2-positive cell-related diseases comprising: administering an effective amount of the fusion protein to a subject in need of alleviating or treating the disease, or a protein expressing the fusion protein An expression system or its culture, or a pharmaceutical composition containing the fusion protein.
  • Figure 1 C57BL/6 mice were single-administered 1mg/kg DR303X2 and 1mg/kg DR303X8 by tail vein injection, and the relationship between blood drug concentration and administration time.
  • Figure 4 The anti-tumor efficacy of anti-CLD18A2/HSA/CD3 trispecific fusion protein in PBMC immune system humanized NUGC4/hCLDN18.2 subcutaneous xenograft model of gastric cancer.
  • a fusion protein with three-target targeting which included: (1) a single-domain antibody (VHH) specifically binding to the CLD18A2 antigen, (2) a CD3-specific Bound single chain antibody (scFv) and (3) single domain antibody (VHH) that specifically binds human serum albumin (HSA).
  • VHH single-domain antibody
  • scFv CD3-specific Bound single chain antibody
  • HSA human serum albumin
  • the anti-CLD18A2 single-domain antibody had poor stability in vivo, and the application of IgG Fc, which is commonly used in the field to improve expression and stability, to be fused to it still cannot achieve substantial improvement.
  • the inventors fused the anti-CLD18A2 domain with the anti-CD3 domain and the anti-HSA domain to obtain an anti-CLD18A2/HSA/CD3 trispecific fusion protein.
  • anti-CLD18A2/HSA/CD3 trispecific fusion protein As used herein, the "anti-CLD18A2/HSA/CD3 trispecific fusion protein”, “trispecific fusion protein”, and “triple target-targeting fusion protein” can be used interchangeably.
  • the term "specificity" in the present invention means that the trispecific fusion protein of the present invention does not or substantially does not or less cross-reacts with other proteins other than the target antigen.
  • the degree of its specificity can be judged by immunological techniques, including but not limited to western blotting, immunoaffinity chromatography, flow cytometry and the like.
  • the specific recognition is preferably determined by flow cytometry, and the criteria for specific recognition in specific cases can be judged by those of ordinary skill in the art based on their common knowledge in the field.
  • the anti-CLD18A2 domain is a single domain antibody, referred to herein as "anti-CLD18A2VHH".
  • the anti-HSA domain is a single domain antibody, which is referred to as “anti-HSA VHH” for short herein.
  • the overall molecular weight of the single-domain antibody provided by the present invention can be about one-half that of the ScFv single-chain antibody, so the molecular weight of the overall structure can be effectively reduced, thereby enhancing its tissue penetration and reaching target tissues and organs more effectively , improve the therapeutic effect, and this structure is easier to prepare than the structure with two ScFvs in series.
  • the anti-CD3 structural domain is a single-chain antibody, which is referred to as "anti-CD3 scFv" for short in the present invention.
  • the inventors have optimized and transformed the anti-CLD18A2 domain and the anti-HSA domain, the FR1 region of the anti-CLD18A2 domain undergoes a P ⁇ A mutation; the FR1 region of the anti-HSA domain undergoes a mutation P ⁇ A mutation.
  • the mutations very significantly increased the stability and half-life of the fusion protein.
  • the anti-CD3 domain is a single-chain antibody, and preferably its amino acid sequence is shown in positions 254-502 of SEQ ID NO:1.
  • the anti-CLD18A2 domain can specifically bind to the CLD18A2 antigen in tumor tissue; the anti-HSA domain can reversibly and non-covalently bind to albumin in serum; the anti-CD3 scFv can bind to T
  • the CD3 subunit in the cellular TCR complex binds, activates T cells, and releases cytokines to kill tumor target cells.
  • sequence (N-terminal-C-terminal) of each domain in the anti-CLD18A2/HSA/CD3 trispecific fusion protein can be: anti-CLD18A2VHH-anti-HSA VHH-anti-CD3scFv or anti-CLD18A2VHH-anti-CD3scFv-anti-HSA VHH.
  • the anti-CLD18A2/HSA/CD3 trispecific fusion protein In the absence of tumor cells and T cells, the anti-CLD18A2/HSA/CD3 trispecific fusion protein non-covalently binds to HSA in peripheral blood, prolonging its in vivo half-life through the circulating mechanism of HSA binding to FcRn. In the tumor microenvironment, the anti-CLD18A2/HSA/CD3 trispecific fusion protein cross-links with tumor cells and T cells expressing CLD18A2 antigen to produce TDCC effect (T-cell dependent cellular cytotoxicity) to kill tumor cells.
  • T-cell dependent cellular cytotoxicity T-cell dependent cellular cytotoxicity
  • a connecting peptide may also be included.
  • anti-CLD18A2VHH and anti-HSA Linking peptides may be provided between VHHs, and between anti-HSA VHHs and anti-CD3 scFv.
  • the connecting peptide fragment can generally be a flexible polypeptide rich in G, S and/or A (mainly composed of glycine (G), serine (S) and/or alanine (A)) with a suitable length, so that Adjacent protein domains are free to move relative to each other.
  • the amino acid sequence of the connecting peptide fragment may include (GS)n, (GGS)n, (GGSG)n, (GGGS)nA, (GGGGS)nA, (GGGGS)nG, (GGGGA)nA, (GGGGG )nA and other sequences, wherein n is selected from an integer between 1-10.
  • the length of the amino acid sequence of the connecting peptide fragment can be 3-30, 3-4, 4-6, 6-8, 8-10, 10-12, 12-14, 14-16, 16-18, 18-20, 20-22, 22-24, 24-26, 26-28 or 28-30.
  • the fusion protein may also contain a tag protein, and the tag protein is 6 ⁇ His, Fc, Myc, GST, Flag or HA.
  • the tag protein is 6 ⁇ His, and the nucleotide sequence of the tag protein is shown in SEQ ID NO:9.
  • the present invention also includes their homofunctional variants.
  • the difference between these variants with the same function and the preferred structural domains listed in the present invention may be the difference in the amino acid sequence, or the difference in the modified form that does not affect the sequence, or both. It should be understood that the change is more likely or usually occurs in the non-CDR region of the domain, but when considering the preferred mutation scheme of the present invention, the P ⁇ A mutation in the FR1 region corresponding to the anti-CLD18A2 domain remains conservative, corresponding The P ⁇ A mutation in the FR1 region of the anti-HSA domain remains conservative.
  • the present invention includes sequences with high homology with the described structural domains (such as 70% or higher homology with the specific protein sequences listed; preferably 80% or higher homology; more preferably 90% or higher homology, such as 95%, 98% or 99% homology), and have the same function.
  • the present invention also includes one or more (such as 1-20, 1-15, 1-10, 1-8, 1-5, 1-3 or 1-2) amino acid residues A domain formed by substitution, deletion or addition of a group and having the same function.
  • the change is more likely or usually occurs in the non-CDR regions of the domain, but Ala at position 14, Ala at position 143 corresponding to the sequence shown in SEQ ID NO: 1 is conserved.
  • the present invention also provides an isolated polynucleotide encoding the anti-CLD18A2/HSA/CD3 trispecific fusion protein of the present invention
  • the polynucleotide may be RNA, DNA or cDNA, etc.
  • Methods for providing the isolated polynucleotide should be known to those skilled in the art, for example, it can be prepared by automatic DNA synthesis and/or recombinant DNA technology, etc., or it can be isolated from a suitable natural source.
  • the nucleic acid sequence of the isolated polynucleotide may include the sequence shown in SEQ ID NO:4.
  • the present invention also provides an expression vector containing the isolated polynucleotide of the present invention.
  • the method for constructing the expression vector should be known to those skilled in the art.
  • the expression vector can be constructed by methods such as in vitro recombinant DNA technology, DNA synthesis technology, and in vivo recombination technology. More specifically, it can be It is constructed by inserting the isolated polynucleotide into the multiple cloning site of the expression vector.
  • the expression vector in the present invention generally refers to various commercially available expression vectors well known in the art, such as bacterial plasmids, phages, yeast plasmids, plant cell viruses, mammalian cell viruses such as adenovirus, retrovirus or other vectors.
  • the vector may also include one or more regulatory sequences operably linked to the polynucleotide sequence, which may include a suitable promoter sequence.
  • the promoter sequence is usually operably linked to the coding sequence for the amino acid sequence to be expressed.
  • the promoter can be any nucleotide sequence that shows transcriptional activity in the host cell of choice, including mutated, truncated, and hybrid promoters, and can be derived from an extracellular sequence that encodes either homologous or heterologous to the host cell. Or intracellular polypeptide gene acquisition.
  • the regulatory sequences may also include suitable transcription terminator sequences, sequences recognized by a host cell to terminate transcription. A terminator sequence is attached to the 3' end of the nucleotide sequence encoding the polypeptide, and any terminator that is functional in the host cell of choice may be used in the present invention.
  • suitable vectors will contain an origin of replication functional in at least one organism, a promoter sequence, convenient restriction enzyme sites and one or more selectable markers.
  • these promoters may be lac or trp promoters including but not limited to E. coli; bacteriophage lambda PL promoter; eukaryotic promoters including CMV immediate early promoter, HSV thymidine kinase promoter, early and late SV40 promoters , the methanol oxidase promoter of Pichia pastoris and some other known promoters that can control the expression of genes in prokaryotic or eukaryotic cells or their viruses.
  • Marker genes may be used to provide phenotypic traits for selection of transformed host cells, for example, may include but are not limited to dihydrofolate reductase for eukaryotic cell culture, neomycin resistance, and green fluorescent protein (GFP), Or tetracycline or ampicillin resistance for E. coli etc.
  • an enhancer sequence can also be included in the expression vector. If the enhancer sequence is inserted into the vector, the transcription will be enhanced.
  • the enhancer is a cis-acting factor of DNA, usually about There are 10 to 300 base pairs and act on the promoter to enhance the transcription of the gene.
  • the present invention also provides an expression system of an anti-CLD18A2/HSA/CD3 trispecific fusion protein, the expression system contains the expression vector of the present invention or the exogenous polynucleotide of the present invention is integrated in the genome.
  • Any cell suitable for expressing the expression vector can be used as a host cell, for example, the host cell can be a prokaryotic cell, such as a bacterial cell; or a lower eukaryotic cell, such as a yeast cell; or a higher Eukaryotic cells, such as mammalian cells, specifically include but are not limited to Escherichia coli; Streptomyces sp; bacterial cells of Salmonella typhimurium; fungal cells such as yeast, filamentous fungi, plant cells; insect cells of Drosophila S2 or Sf9 ; A combination of one or more of CHO, COS, HEK293 cells, or animal cells such as Bowes melanoma cells.
  • the method for constructing the expression system should be known to those skilled in the art, for example, it may include but not limited to microinjection method, particle gun method, electroporation method, virus-mediated transformation method, electron bombardment method , calcium phosphate precipitation method, etc. in one or more combinations.
  • the expression system is an animal cell expression system, such as CHO, NSO, BHK, HEK-293 or PER-C6. In a more specific embodiment, said cell is CHO.
  • the invention also provides a preparation method of the anti-CLD18A2/HSA/CD3 trispecific fusion protein.
  • a preparation method may include: cultivating the expression system of the fusion protein of the present invention under conditions suitable for expressing the fusion protein, so as to express The fusion protein is obtained, and the fusion protein is purified and isolated.
  • the present invention also provides a pharmaceutical composition, including the anti-CLD18A2/HSA/CD3 trispecific fusion protein of the present invention, the expression system of the present invention or its culture.
  • the pharmaceutically acceptable carrier may include various excipients and diluents, these carriers themselves are not essential active ingredients, and there is no excessive toxicity after administration. Suitable carriers will be well known to those skilled in the art, for example, a full discussion of pharmaceutically acceptable carriers can be found in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J., 1991).
  • the content of the fusion protein is usually an effective amount, and the content of the active ingredient corresponding to the effective amount can be determined according to the object to be treated and the specific administration method.
  • the present invention also provides a kit containing the fusion protein, the expression system or its expression product (culture) or the pharmaceutical composition. Instructions for use can also be included in the kit to guide the application of those skilled in the art or clinicians.
  • the present invention also provides the use of the anti-CLD18A2/HSA/CD3 trispecific fusion protein, the expression system or its culture in the preparation of medicines for alleviating or treating diseases related to CLD18A2 positive cells.
  • CLD18A2-positive cell (related) disease includes “CLD18A2 abnormal expression (related) disease” or “CLD18A2 high expression (related) disease”, and these terms may be Used interchangeably.
  • CLD18A2 is expressed in many tumor cells.
  • CLD18A2 can be used as a therapeutic target for related tumors.
  • the CLD18A2-positive cells may include all CLD18A2-expressing tumors, and these tumors may usually express CLD18A2-positively.
  • the positive expression of CLD18A2 usually refers to the expression of CLD18A2, or the expression level of CLD18A2 is higher than a certain standard.
  • the expression of CLD18A2 protein can be detected.
  • CLD18A2 positive can be that the mRNA expression level of CLD18A2 in the tumor tissue is higher than that of its surrounding healthy tissue.
  • CLD18A2 positive can be that the expression level of CLD18A2 protein in the tumor tissue is higher than its surrounding healthy tissue.
  • diseases may specifically include but are not limited to gastric cancer, esophageal cancer, pancreatic cancer, lung cancer, ovarian cancer, breast cancer, colorectal cancer, liver cancer, gallbladder cancer, head and neck cancer, etc. These cancers may be early, middle or late, such as metastasis cancer etc.
  • the present invention also provides a treatment method comprising: administering to an individual an effective amount of the anti-CLD18A2/HSA/CD3 trispecific fusion protein, the expression system or its culture, or the pharmaceutical composition.
  • the "therapeutically effective amount" of the fusion protein and pharmaceutical composition provided by the present invention preferably leads to a reduction in the severity of disease symptoms, an increase in the frequency and duration of the asymptomatic period of the disease, or prevention of damage or disability caused by the suffering of the disease .
  • a "therapeutically effective amount” preferably inhibits cell growth or tumor growth by at least about 10%, preferably at least about 20%, more preferably at least About 30%, more preferably at least about 40%, more preferably at least about 50%, more preferably at least about 60%, more preferably at least about 70%, more preferably at least about 80%.
  • the ability to inhibit tumor growth can be evaluated in animal model systems predictive of efficacy against human tumors. Alternatively, it can also be assessed by examining the ability to inhibit cell growth, which can be determined in vitro by assays well known to those skilled in the art.
  • a therapeutically effective amount of a fusion protein, pharmaceutical composition generally reduces tumor size, or otherwise alleviates symptoms in a subject. Those skilled in the art can select an appropriate therapeutically effective amount according to the actual situation, for example, the size of the subject, the severity of the subject's symptoms and the selected specific composition or administration route.
  • Prescriptions for treatment can be determined by a physician, generally taking into account factors including, but not limited to, the disease being treated, individual patient conditions, site of delivery, method of administration, and other factors.
  • Prophylactically effective dose refers to the amount that is effective in achieving the desired preventive effect at the necessary dose and time. quantity.
  • the fusion protein and pharmaceutical composition of the present invention can be administered as a single active ingredient, or in combination therapy, that is, in combination with other agents.
  • the combination therapy may be the fusion protein or the pharmaceutical composition combined with at least one other anti-tumor drug (for example, paclitaxel, etc.).
  • the combination therapy may be the combination of the fusion protein, the pharmaceutical composition and an immune checkpoint inhibitor, and the immune checkpoint inhibitor includes but is not limited to a PD-1 inhibitor, a PD-L1 inhibitor, or A combination of one or more of CTLA-4 inhibitors, etc., said inhibitors may be monoclonal antibodies.
  • MOLECULAR CLONING A LABORATORY MANUAL, Second edition, Cold Spring Harbor Laboratory Press, 1989 and Third edition, 2001; Ausubel et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons , New York, 1987 and periodic updates; the series METHODS IN ENZYMOLOGY, Academic Press, San Diego; Wolffe, CHROMATIN STRUCTURE AND FUNCTION, Third edition, Academic Press, San Diego, 1998; METHODS IN ENZYMOLOGY, Vol.304, Ch romatin (PM Wassarman and AP Wolffe, eds.), Academic Press, San Diego, 1999; and METHODS IN MOLECULAR BIOLOGY, Vol.119, Chromatin Protocols (PB Becker, ed.) Humana Press, Totowa, 1999 et al.
  • Embodiment 1 the fusion expression of trispecific fusion protein
  • Positions 1-120 are anti-CLDN18.2 VHH, positions 130-244 are anti-HSA VHH, positions 254-502 are anti-CD3 scFv.
  • Amino acids underlined in italics are linker sequences.
  • Amino acid fragments marked in bold underline are CDRs.
  • the 26th to 33rd positions are CDR1
  • the 51st to 57th positions are CDR2
  • the 96th to 109th positions are CDR3.
  • positions 155-162 are CDR1
  • positions 178-187 are CDR2
  • positions 226-233 are CDR3.
  • the 279th-286th position is HCDR1
  • the 304th-313rd position is HCDR2
  • the 352th-367th position is HCDR3
  • the 419th-428th position is LCDR1
  • the 445th-447th position is LCDR2
  • the 484th-492nd position is LCDR3.
  • the CDR region was marked using the IMGT numbering scheme (antibody numbering scheme).
  • positions 1-120 are anti-CLDN18.2 VHH; positions 130-378 are anti-CD3 scFv (excluding anti-HSA VHH).
  • Amino acid fragments marked in bold underline are CDRs.
  • the 26th to 33rd positions are CDR1
  • the 51st to 57th positions are CDR2
  • the 96th to 109th positions are CDR3.
  • 155th to 162th is HCDR1
  • 180th to 189th is HCDR2
  • 228th to 243rd is HCDR3
  • Bits 295 to 303 are LCDR1
  • bits 321 to 323 are LCDR2
  • bits 360 to 368 are LCDR3.
  • the CDR region was marked using the IMGT numbering scheme (antibody numbering scheme).
  • positions 1-120 are anti-CLDN18.2 VHH, positions 130-244 are anti-HSA VHH, positions 254-502 are anti-CD3 scFv.
  • Amino acids underlined in italics are linker sequences.
  • Amino acid fragments marked in bold underline are CDRs.
  • the 26th to 33rd positions are CDR1
  • the 51st to 57th positions are CDR2
  • the 96th to 109th positions are CDR3.
  • positions 155-162 are CDR1
  • positions 178-187 are CDR2
  • positions 226-233 are CDR3.
  • the 279th-286th position is HCDR1
  • the 304th-313rd position is HCDR2
  • the 352th-367th position is HCDR3
  • the 419th-428th position is LCDR1
  • the 445th-447th position is LCDR2
  • the 484th-492nd position is LCDR3.
  • the CDR region was marked using the IMGT numbering scheme (antibody numbering scheme).
  • the anti-CLD18A2 domain, anti-CD3 domain and anti-human serum albumin (HSA) domain were combined in tandem through connecting peptides to form an anti-CLD18A2/HSA/CD3 trispecific fusion protein (Table 1), and entrusted to General Biosystems ( Anhui) Co., Ltd. optimized the synthesis of the base sequence. After adding a signal peptide (METDTLLLWVLLLWVPGSTG) at the 5' end, it was constructed into an animal cell expression vector for secreted expression.
  • X910 According to the sequence of SEQ ID NO: 132 of US_2020_0055932_A1, it was expressed and prepared in CHO cells.
  • the cell expression supernatant was centrifuged at high speed, then filtered through a 1.2 ⁇ m membrane, and loaded onto an AT Protein A Diamond affinity chromatography column (Borgeron (Shanghai) Biotechnology Co., Ltd.).
  • Equilibrium solution is 20mmol/L Tris-HCl, 150mmol/L NaCl, pH7.4 solution; eluent is 20mmol/L HAc-NaAc pH5.0 solution; eluent is 100mmol/L Gly-HCl, pH3.0 solution ;
  • the cleaning solution is 0.1mmol/L NaOH solution; the neutralizing solution is 1mol/L Tris-HCl, pH8.5.
  • the anion-exchange flow-through sample was loaded onto a Diamond SP cation-exchange chromatography column (Borgeron (Shanghai) Biotechnology Co., Ltd.), 0-100% B linear gradient elution (0-500mmol/L NaCl, 20mmol/L PB , pH7.0 solution, 20CV), obtain SDS-PAGE electrophoresis purity ⁇ 95% sample.
  • Embodiment 2 in vitro activity detection
  • CHO-C18.2-gfpx stably transfected cell line target cell highly expressing CLDN18A2: CLD18A2 full-length gene (sequence shown in SEQ ID NO: 7) and egfp gene (sequence shown in SEQ ID NO: 8 Shown) double expression cassette plasmid pDR05-C18.2-gfp, the plasmid was extracted with an endotoxin-free plasmid kit, electroporated into CHO-S cells after sterile filtration, and cultured in 96-well plates with MSX under pressure.
  • Jurkat-NFAT-Luc2p effector cell stably transfected cell line: Jurkat cells were transformed into the NFAT-responsive luciferase system to obtain the TDCC activity detection effector cell line Jurkat-NFAT-Luc2p.
  • the protein was diluted to the specified concentration with RPMI1640 medium (containing 1% FBS), and then an appropriate volume (25 ⁇ l/well) was added to a 96-well plate, and cultured overnight for 18 hours. Add 10 ⁇ l/well of detection solution (luciferase detection kit, Promega, E2620), shake for 1.5 min, transfer 60 ⁇ l of the solution to a white plate, and detect in a microplate reader (Molecular Devices I3).
  • detection solution luciferase detection kit, Promega, E2620
  • Example 2 The reporter gene method in Example 2 was used to compare the stability of DR303X2 and DR303X8 incubated in human serum for 7 days at 37°C.
  • CHO-C18.2-gfpx and Jurkat-NFAT-Luc2p-10F2 cells centrifuge at 1000rpm for 5/10min, discard the supernatant, add 1640 medium (containing 1% FBS) to resuspend, count, and use the analysis medium to adjust Effector cell Jurkat-NFAT-luc2p-10F2 cell density to 2.4 ⁇ 10 6 cells/ml, adjust target cell CHO-C18.2-gfpx cell density to 6 ⁇ 10 5 cells/ml, mix 1:1 and spread in 96-well plate , 50 ⁇ l per well, spare.
  • 1640 medium containing 1% FBS
  • detection solution luciferase detection kit, Promega, E2620
  • the TDCC activity of DR303X2 and DR303X8 samples in human serum showed a downward trend with increasing storage time at 37°C.
  • the EC 50 of the DR303X2 sample in human serum (HS) was 0.04012 on day 0, and the EC 50 on day 7 was 0.07349, and the residual activity rate was 55%.
  • the EC 50 of the DR303X8 sample in human serum (HS) was 0.01426 at day 0, and the EC 50 at day 7 was 0.02265, and the residual activity rate was 63%.
  • Activity residual rate EC 50 (0 day) /EC 50 (7 day) *100%.
  • DR303X8 has better stability in human serum than DR303X2.
  • Embodiment 4 half-life in vivo of C57BL/6 mice
  • C57BL/6 mice were given 1 mg/kg DR303X2 and 1 mg/kg DR303X8 by tail vein injection, and blood samples were collected 3 days before the drug, 1 h, 6 h, 24 h, 30 h, 48 h, 72 h, 96 h, and 120 h after the drug to analyze the drug concentration.
  • the pharmacokinetic parameters were calculated using the non-compartmental model using PK solver 2.0 software.
  • the half-lives were 26.8h and 28.8h respectively.
  • Embodiment 5 half-life in cynomolgus monkeys
  • Cynomolgus monkeys were given 0.03mg/kg, 0.6mg/kg, 3mg/kg DR303X8 or DR303X6 intravenously in a single dose, and the blood was sampled and analyzed before administration, immediately after administration, 4h, 8h, 24h, 48h, 72h, 120h and 168h Drug concentrations were calculated, and pharmacokinetic parameters were calculated using non-compartmental models.
  • DR303X8 or DR303X6 was intravenously injected into cynomolgus monkeys: when the dose was 0.03mg/kg, the C max was 1.02 ⁇ 0.02 ⁇ g/ml, the AUC 0-t was 33.12 ⁇ 0.12 ⁇ g/ml*h, and the t 1/2 was 46.03 ⁇ 16.32h; when the dose was 0.6mg/kg, C max was 17.33 ⁇ 0.65 ⁇ g/ml, AUC 0-t was 641.74 ⁇ 102.66 ⁇ g/ml*h, t 1/2 was 37.37 ⁇ 1.11h; At 3mg/kg, C max was 81.01 ⁇ 2.44 ⁇ g/ml, AUC 0-t was 3846.04 ⁇ 497.80 ⁇ g/ml*h, t 1/2 was 41.8 ⁇ 5.76h.
  • DR303X8 has a long half-life in vivo and ideal stability in vivo.
  • Example 6 Anti-tumor efficacy of anti-CLD18A2/HSA/CD3 trispecific fusion protein in PBMC immune system humanized BxPC3/hCLDN18.2 pancreatic cancer subcutaneous xenograft tumor model
  • BxPC3/hCLDN18.2 pancreatic cancer subcutaneous xenograft model To establish a PBMC immune system humanized BxPC3/hCLDN18.2 pancreatic cancer subcutaneous xenograft model, select male NCG mice, about 7 weeks old. BxPC3/hCLDN18.2 tumor cells were inoculated subcutaneously in the right flank of mice, 5x10 6 /mouse, PBMCs were derived from normal human peripheral blood, and injected into tumor-bearing mice by tail vein, 2x10 6 /mouse.
  • the drugs were divided into groups, divided into 5 groups, 6 animals in each group, and blank preparation (vehicle, 2 times a week, continuous administration for 5 weeks), 0.03mg/kg DR303X8 (twice a week, continuous administration for 5 weeks), 0.03mg/kg DR303X8 (once a day, continuous administration for 17 days), 0.1mg/kg DR303X8 (twice a week, continuous administration for 5 weeks), 0.1mg /kg DR303X8 (once a day, continuous administration of 17 Days), drug withdrawal and observation.
  • Example 7 Anti-tumor efficacy of anti-CLD18A2/HSA/CD3 trispecific fusion protein in PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft tumor model
  • the PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft model was established, and male NCG mice, about 7 weeks old, were selected.
  • NUGC4/hCLDN18.2 tumor cells were inoculated subcutaneously in the right flank of mice, 5x10 6 /mouse, PBMCs were derived from normal human peripheral blood, and injected into tumor-bearing mice by tail vein, 2x10 6 /mouse.
  • the drugs were divided into groups, divided into 5 groups, 6 animals in each group, and blank preparation (vehicle, 2 times a week, continuous administration for 5 weeks), 0.03mg/kg DR303X8 (twice a week, continuous administration for 5 weeks), 0.03mg/kg DR303X8 (once a day, continuous administration for 17 days), 0.1mg/kg DR303X8 (twice a week, continuous administration for 5 weeks), 0.1mg /kg DR303X8 (once a day, continuous administration for 17 days).
  • Example 8 Antitumor efficacy of fusion proteins DR303X8 and X910 in PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft tumor model
  • the PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft model was selected, and male NCG mice were selected, about 7 weeks old.
  • NUGC4/hCLDN18.2 tumor cells were inoculated subcutaneously in the right flank of mice, 5x10 6 /mouse;
  • PBMCs were derived from normal human peripheral blood, and injected into tumor-bearing mice by tail vein, 2x10 6 /mouse.
  • the drugs were divided into groups, divided into 5 groups, with 6 animals in each group, and the blank preparation (vehicle, once a day, continuously administered for 29 days), 0.01 mg/kg DR303X8 ( Once a day, continuous administration for 29 days), 0.03mg/kg DR303X8 (twice a week, continuous administration for 5 times), 0.03mg/kg DR303X8 (once a day, continuous administration for 17 days), 0.06mg/kg X910 (once a day, continuous administration for 32 days).
  • DR303X8 has a significant tumor inhibitory effect on the PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft tumor model.

Abstract

A three-target anti-tumor drug, and a preparation method therefor and the use thereof. A fusion protein can be efficiently expressed, has a long half-life in vivo, a high stability and a good tumor inhibitory effect.

Description

一种三靶点抗肿瘤药物、其制备方法及其应用A kind of antitumor drug with three targets, its preparation method and application 技术领域technical field
本发明涉及生物领域,特别是涉及一种特异性靶向于CLD18A2、人血清白蛋白和CD3的三靶点抗肿瘤药物及其制备方法和用途。The invention relates to the field of biology, in particular to a three-target antitumor drug specifically targeting CLD18A2, human serum albumin and CD3, its preparation method and application.
背景技术Background technique
密蛋白18(Claudin 18,CLD18)是分子量约为28kD,位于上皮和内皮的紧密连接中的跨膜蛋白,紧密连接在相邻细胞之间。在正常上皮组织中,由于细胞间隙紧密导致细胞表面的密蛋白难以接触,而肿瘤细胞的间隙却较为疏松,因此肿瘤细胞上的密蛋白成为胞外抗体及免疫疗法的潜在靶点。CLD18具有四个疏水区,其作为跨膜区形成两个胞外域,其中疏水区1和疏水区2环绕形成胞外域1,疏水区3和疏水区4环绕形成胞外域2。人的CLD18基因具有两个不同的1号外显子,转录后经过可变剪接最终生成仅在N端具有不同序列的两个蛋白亚型:CLD18A1和CLD18A2。CLD18A1在正常肺组织中表达,而CLD18A2仅在胃组织中表达;更重要的是,CLD18A2局限在已分化的胃上皮短寿细胞中,但在胃干细胞中不存在(Niimi T,et al.Biol.2001;21(21):7380-7390)。Claudin 18 (CLD18) is a transmembrane protein with a molecular weight of about 28kD, located in the tight junction of epithelium and endothelium, and is tightly connected between adjacent cells. In normal epithelial tissue, claudin on the cell surface is difficult to access due to the tight intercellular space, while the intercellular space in tumor cells is relatively loose. Therefore, claudin on tumor cells has become a potential target for extracellular antibodies and immunotherapy. CLD18 has four hydrophobic regions, which form two extracellular domains as transmembrane regions, in which hydrophobic region 1 and hydrophobic region 2 surround to form extracellular domain 1, and hydrophobic region 3 and hydrophobic region 4 surround to form extracellular domain 2. The human CLD18 gene has two different exon 1, which can be alternatively spliced after transcription to generate two protein isoforms with different sequences only at the N-terminus: CLD18A1 and CLD18A2. CLD18A1 is expressed in normal lung tissue, whereas CLD18A2 is expressed only in gastric tissue; more importantly, CLD18A2 is restricted to differentiated short-lived gastric epithelial cells but absent from gastric stem cells (Niimi T, et al. Biol .2001;21(21):7380-7390).
此外,CLD18A2在原发性胃癌及其转移后癌症类型中存在高表达,在胰腺癌,食管癌,卵巢癌,肺癌中也常常观察到其被激活表达。CLD18A2的这些特性使其成为临床上治疗胃癌和其他相关肿瘤的潜在靶点。In addition, CLD18A2 is highly expressed in primary gastric cancer and its metastatic cancer types, and its activated expression is often observed in pancreatic cancer, esophageal cancer, ovarian cancer, and lung cancer. These properties of CLD18A2 make it a potential target for clinical treatment of gastric cancer and other related tumors.
然而,虽然目前已有数个针对CLD18A2靶点的药物处于临床阶段,但是仍未有获批上市的药物。现有的针对CLD18A2靶点的药物研究中,在药物表达系统、药物体内半衰期、药效以及靶向性等方面,还存在有待提高的方面。However, although several drugs targeting CLD18A2 are currently in the clinical stage, there are still no drugs approved for marketing. In the existing drug research on the CLD18A2 target, there are still aspects to be improved in terms of drug expression system, drug half-life in vivo, drug efficacy, and targeting.
发明内容Contents of the invention
本发明的目的在于提供一种特异性靶向CLD18A2、人血清白蛋白和CD3的三靶点抗肿瘤药物及其制备方法和用途。The object of the present invention is to provide a three-target anti-tumor drug specifically targeting CLD18A2, human serum albumin and CD3, its preparation method and application.
在本发明的第一方面,提供一种融合蛋白,包括:抗CLD18A2结构域、抗人血清白蛋白(HSA)结构域和抗CD3结构域;所述抗CLD18A2结构域为单域抗体,具有SEQ ID NO:1中第26-33位所示的CDR1,SEQ ID NO:1中第51-57位所示的CDR2,SEQ ID NO:1中第96-109位所示的CDR3;所述抗人血清白蛋白结构域为单域抗体,具有SEQ ID NO:1中第155-162位所示的CDR1,SEQ ID NO:1中第180-187位所示的CDR2,SEQ ID NO:1中第226-233位所示的CDR3。 In a first aspect of the present invention, a fusion protein is provided, comprising: an anti-CLD18A2 domain, an anti-human serum albumin (HSA) domain and an anti-CD3 domain; the anti-CLD18A2 domain is a single-domain antibody with SEQ ID NO: CDR1 shown in No. 26-33 in 1, CDR2 shown in No. 51-57 in SEQ ID NO: 1, CDR3 shown in No. 96-109 in SEQ ID NO: 1; Said anti The human serum albumin domain is a single domain antibody with CDR1 shown in positions 155-162 in SEQ ID NO:1, CDR2 shown in positions 180-187 in SEQ ID NO:1, and in SEQ ID NO:1 CDR3 indicated at positions 226-233.
在一种或多种实施方式中,所述抗CLD18A2结构域为单域抗体,其框架区(FR)存在氨基酸序列的P→A突变;较佳地,其FR1区存在P→A突变;更佳地,其P→A突变对应SEQ ID NO:1序列的第14位。In one or more embodiments, the anti-CLD18A2 domain is a single domain antibody, and its framework region (FR) has a P→A mutation in the amino acid sequence; preferably, its FR1 region has a P→A mutation; more Preferably, the P→A mutation corresponds to the 14th position of SEQ ID NO:1.
在一种或多种实施方式中,所述抗人血清白蛋白结构域为单域抗体,其框架区(FR)存在氨基酸序列的P→A突变;较佳地,其FR1区存在P→A突变;更佳地,其P→A突变的氨基酸为对应SEQ ID NO:1序列的第143位的氨基酸。In one or more embodiments, the anti-human serum albumin domain is a single domain antibody, and its framework region (FR) has a P→A mutation in its amino acid sequence; preferably, its FR1 region has a P→A mutation Mutation; more preferably, the amino acid of its P→A mutation is the 143rd amino acid corresponding to the sequence of SEQ ID NO:1.
在一种或多种实施方式中,所述抗CD3结构域为单链抗体。In one or more embodiments, the anti-CD3 domain is a single chain antibody.
在一种或多种实施方式中,所述抗CD3结构域具有SEQ ID NO:1中第279-286位所示的HCDR1,SEQ ID NO:1中第304-313位所示的HCDR2,SEQ ID NO:1中第352-367位所示的HCDR3,SEQ ID NO:1中第419-427位所示的LCDR1,SEQ ID NO:1中第445-447位所示的LCDR2,SEQ ID NO:1中第484-492位所示的LCDR3。In one or more embodiments, the anti-CD3 domain has HCDR1 shown in positions 279-286 in SEQ ID NO: 1, HCDR2 shown in positions 304-313 in SEQ ID NO: 1, SEQ ID NO: 1 HCDR3 shown in No. 352-367 in ID NO:1, LCDR1 shown in No. 419-427 in SEQ ID NO:1, LCDR2 shown in No. 445-447 in SEQ ID NO:1, SEQ ID NO : LCDR3 shown in bits 484-492 in 1.
在一种或多种实施方式中,所述的抗CD3结构域位于抗人血清白蛋白结构域是N端或C端。In one or more embodiments, the anti-CD3 domain is located at the N-terminal or C-terminal of the anti-human serum albumin domain.
在一种或多种实施方式中,所述抗CLD18A2结构域的氨基酸序列如SEQ ID NO:1中第1~120位所示。In one or more embodiments, the amino acid sequence of the anti-CLD18A2 domain is shown in positions 1-120 of SEQ ID NO:1.
在一种或多种实施方式中,所述抗人血清白蛋白结构域的氨基酸序列如SEQ ID NO:1中第130~244位所示。In one or more embodiments, the amino acid sequence of the anti-human serum albumin domain is shown in positions 130-244 of SEQ ID NO:1.
在一种或多种实施方式中,所述抗CD3结构域的氨基酸序列如SEQ ID NO:1中第254~502位所示。In one or more embodiments, the amino acid sequence of the anti-CD3 domain is shown in positions 254-502 of SEQ ID NO:1.
在一种或多种实施方式中,所述抗CLD18A2结构域、抗人血清白蛋白结构域和抗CD3结构域的氨基酸序列直接连接,或通过连接子(连接肽)连接;较佳地,所述的融合蛋白的氨基酸序列如SEQ ID NO:1所示。In one or more embodiments, the amino acid sequences of the anti-CLD18A2 domain, the anti-human serum albumin domain and the anti-CD3 domain are directly connected, or connected through a linker (connecting peptide); preferably, the The amino acid sequence of the fusion protein is shown in SEQ ID NO:1.
在一种或多种实施方式中,所述的连接子包括但不限于:含有甘氨酸(G)、丝氨酸(S)和/或丙氨酸(A)的柔性多肽。In one or more embodiments, the linker includes, but is not limited to: a flexible polypeptide containing glycine (G), serine (S) and/or alanine (A).
在一种或多种实施方式中,当通过两个连接子连接时,所述两个连接子相同;或者,所述两个连接子不同。In one or more embodiments, when linked by two linkers, the two linkers are the same; alternatively, the two linkers are different.
在一种或多种实施方式中,所述的连接子例如为2A,如P2A,E2A,T2A。In one or more embodiments, the linker is, for example, 2A, such as P2A, E2A, T2A.
在一种或多种实施方式中,所述的抗CLD18A2VHH、抗HSA VHH与抗CD3scFv,从N端到C端依次为:CLD18A2VHH、抗HSA VHH和抗CD3scFv。In one or more embodiments, the anti-CLD18A2 VHH, anti-HSA VHH and anti-CD3 scFv, from N-terminus to C-terminus, are: CLD18A2VHH, anti-HSA VHH and anti-CD3 scFv.
在一种或多种实施方式中,所述的抗CLD18A2VHH、抗HSA VHH与抗CD3scFv,从N端到C端依次为:CLD18A2VHH、抗CD3scFv和抗HSA VHH。In one or more embodiments, the anti-CLD18A2 VHH, anti-HSA VHH and anti-CD3 scFv, from N-terminus to C-terminus, are: CLD18A2VHH, anti-CD3 scFv and anti-HSA VHH.
在本发明的另一方面,提供一种多核苷酸或含有该多核苷酸的表达构建体(如 表达载体),所述多核苷酸编码所述的融合蛋白。In another aspect of the present invention, there is provided a polynucleotide or an expression construct comprising the polynucleotide (such as expression vector), the polynucleotide encodes the fusion protein.
在本发明的另一方面,提供一种融合蛋白的表达系统,所述表达系统含有所述的表达构建体或其基因组中整合有外源的所述的多核苷酸。In another aspect of the present invention, a fusion protein expression system is provided. The expression system contains the expression construct or the exogenous polynucleotide integrated in its genome.
在一种或多种实施方式中,所述的表达系统为宿主细胞。In one or more embodiments, the expression system is a host cell.
在一种或多种实施方式中,所述的表达系统为真核表达系统。In one or more embodiments, the expression system is a eukaryotic expression system.
在一种或多种实施方式中,所述的真核表达系统为动物细胞表达系统或酵母表达系统。In one or more embodiments, the eukaryotic expression system is an animal cell expression system or a yeast expression system.
在一种或多种实施方式中,所述的动物细胞表达系统包括(但不限于):CHO,NS0,BHK,HEK-293或PER-C6。In one or more embodiments, the animal cell expression system includes (but not limited to): CHO, NSO, BHK, HEK-293 or PER-C6.
在一种或多种实施方式中,所述的动物细胞表达系统为CHO。In one or more embodiments, the animal cell expression system is CHO.
在一种或多种实施方式中,所述的表达系统为原核表达系统,如原核宿主细胞。In one or more embodiments, the expression system is a prokaryotic expression system, such as a prokaryotic host cell.
在本发明的另一方面,提供制备所述的融合蛋白的方法,包括:利用所述的表达系统表达所述的融合蛋白;较佳地,所述方法还包括:分离出所述的融合蛋白。In another aspect of the present invention, there is provided a method for preparing the fusion protein, comprising: using the expression system to express the fusion protein; preferably, the method further includes: isolating the fusion protein .
在本发明的另一方面,提供一种改造CLD18A2结合分子以提高其疗效的方法,包括:以抗CLD18A2结构域作为CLD18A2结合分子,将其与抗人血清白蛋白结构域和抗CD3结构域连接;所述抗CLD18A2结构域为单域抗体,具有SEQ ID NO:1中第26-33位所示的CDR1,SEQ ID NO:1中第51-57位所示的CDR2,SEQ ID NO:1中第96-109位所示的CDR3;所述抗人血清白蛋白结构域为单域抗体,具有SEQ ID NO:1中第155-162位所示的CDR1,SEQ ID NO:1中第180-187位所示的CDR2,SEQ ID NO:1中第226-233位所示的CDR3。In another aspect of the present invention, a method for modifying a CLD18A2 binding molecule to improve its therapeutic effect is provided, comprising: using an anti-CLD18A2 domain as a CLD18A2 binding molecule, and linking it to an anti-human serum albumin domain and an anti-CD3 domain ; The anti-CLD18A2 structural domain is a single domain antibody, with CDR1 shown in the 26th-33rd position in SEQ ID NO: 1, CDR2 shown in the 51-57th position in SEQ ID NO: 1, SEQ ID NO: 1 The CDR3 shown in the 96th-109th position; the anti-human serum albumin domain is a single domain antibody, with the CDR1 shown in the 155th-162th position in SEQ ID NO:1, and the 180th position in SEQ ID NO:1 - CDR2 shown in position 187, CDR3 shown in positions 226-233 in SEQ ID NO:1.
在本发明的另一方面,提供前面任一所述的融合蛋白、所述的表达系统或其培养物在制备缓解或治疗CLD18A2阳性细胞相关疾病的药物中的用途;较佳地,所述CLD18A2阳性细胞相关疾病包括肿瘤;较佳地,所述肿瘤包括(但不仅限于):胰腺癌、胃癌、食管癌、肺癌、卵巢癌、乳腺癌、结肠直肠癌、肝癌、胆囊癌或头颈癌。In another aspect of the present invention, there is provided the use of any of the aforementioned fusion proteins, the expression system or its culture in the preparation of drugs for alleviating or treating diseases related to CLD18A2 positive cells; preferably, the CLD18A2 Positive cell-related diseases include tumors; preferably, the tumors include (but not limited to): pancreatic cancer, gastric cancer, esophageal cancer, lung cancer, ovarian cancer, breast cancer, colorectal cancer, liver cancer, gallbladder cancer or head and neck cancer.
在本发明的另一方面,提供一种药物组合物,其中包括:所述的融合蛋白。In another aspect of the present invention, a pharmaceutical composition is provided, which includes: said fusion protein.
在本发明的另一方面,提供一种药物组合物,其中所述的融合蛋白的表达系统,或其培养物。In another aspect of the present invention, there is provided a pharmaceutical composition, wherein said fusion protein expression system, or its culture.
在本发明的另一方面,提供一种药盒,其中包括:容器或包装,以及位于其中的所述的融合蛋白。In another aspect of the present invention, a kit is provided, which includes: a container or package, and the fusion protein therein.
在本发明的另一方面,提供一种药盒,其中包括:容器或包装,以及位于其中的所述的融合蛋白的表达系统,或其培养物。In another aspect of the present invention, a kit is provided, which includes: a container or package, and the expression system of the fusion protein or its culture located therein.
在本发明的另一方面,提供一种药盒,其中包括:容器或包装,以及位于其 中的所述的药物组合物。In another aspect of the present invention, a kit is provided, comprising: a container or package, and a The pharmaceutical composition described in.
在本发明的另一方面,提供一种缓解或治疗CLD18A2阳性细胞相关疾病的方法,所述方法包括:给予需要缓解或治疗疾病的对象有效量的所述的融合蛋白,或表达该融合蛋白的表达系统或其培养物,或含有该融合蛋白的药物组合物。In another aspect of the present invention, a method for alleviating or treating CLD18A2-positive cell-related diseases is provided, the method comprising: administering an effective amount of the fusion protein to a subject in need of alleviating or treating the disease, or a protein expressing the fusion protein An expression system or its culture, or a pharmaceutical composition containing the fusion protein.
本发明的其它方面由于本文的公开内容,对本领域的技术人员而言是显而易见的。Other aspects of the invention will be apparent to those skilled in the art from the disclosure herein.
附图说明Description of drawings
图1C57BL/6小鼠尾静脉注射单次给予1mg/kg DR303X2和1mg/kg DR303X8,血药浓度与给药时间关系图。Figure 1 C57BL/6 mice were single-administered 1mg/kg DR303X2 and 1mg/kg DR303X8 by tail vein injection, and the relationship between blood drug concentration and administration time.
图2、DR303X8静脉注射,在高中低剂量组给药浓度(3mg/kg、0.6mg/kg、0.03mg/kg)比为100:20:1时,Cmax比为80:17:1,AUC0-t比为116:20:1,系统暴露量与给药剂量线性相关。Figure 2. Intravenous injection of DR303X8, when the administration concentration (3mg/kg, 0.6mg/kg, 0.03mg/kg) ratio of 100:20:1 in the high, middle and low dose groups, the C max ratio is 80:17:1, AUC The 0-t ratio was 116:20:1, and the systemic exposure was linearly related to the administered dose.
图3、抗CLD18A2/HSA/CD3三特异性融合蛋白在PBMC免疫系统人源化BxPC3/hCLDN18.2胰腺癌皮下移植瘤模型中的的抑瘤药效。biwX5表示每周给药2次、共给药5周;qd X17表示每日给药1次、给药17天;后图中依此类推。Figure 3. Anti-tumor efficacy of anti-CLD18A2/HSA/CD3 trispecific fusion protein in PBMC immune system humanized BxPC3/hCLDN18.2 pancreatic cancer subcutaneous xenograft model. biwX5 means administration twice a week for a total of 5 weeks; qd X17 means administration once a day for 17 days; and so on in the following figure.
图4、抗CLD18A2/HSA/CD3三特异性融合蛋白在PBMC免疫系统人源化NUGC4/hCLDN18.2胃癌皮下移植瘤模型中的的抑瘤药效。Figure 4. The anti-tumor efficacy of anti-CLD18A2/HSA/CD3 trispecific fusion protein in PBMC immune system humanized NUGC4/hCLDN18.2 subcutaneous xenograft model of gastric cancer.
图5、融合蛋白DR303X8和X910在PBMC免疫系统人源化NUGC4/hCLDN18.2胃癌皮下移植瘤模型中的的抑瘤药效。Figure 5. Antitumor efficacy of fusion proteins DR303X8 and X910 in PBMC immune system humanized NUGC4/hCLDN18.2 subcutaneous xenograft model of gastric cancer.
具体实施方式Detailed ways
本发明人经过深入的研究,提供了一种具有三靶点靶向性的融合蛋白,其包括:(1)与CLD18A2抗原特异性结合的单域抗体(VHH)、(2)与CD3特异性结合的单链抗体(scFv)和(3)与人血清白蛋白(HSA)特异性结合的单域抗体(VHH)。所述融合蛋白能够被高效地表达,在体内半衰期长、稳定性高,且具有良好的肿瘤抑制作用。After in-depth research, the inventors provided a fusion protein with three-target targeting, which included: (1) a single-domain antibody (VHH) specifically binding to the CLD18A2 antigen, (2) a CD3-specific Bound single chain antibody (scFv) and (3) single domain antibody (VHH) that specifically binds human serum albumin (HSA). The fusion protein can be efficiently expressed, has a long half-life in vivo, high stability, and has good tumor suppressing effect.
融合蛋白fusion protein
在本发明人的前期研究中,发现抗CLD18A2的单域抗体在体内的稳定性差,而应用IgG Fc此类本领域中常用于改善表达量和稳定性的结构域与之融合仍然无法实现实质上的改善。经过广泛研究筛选,本发明人将抗CLD18A2结构域与抗CD3结构域和抗HSA结构域共同融合,获得一种抗CLD18A2/HSA/CD3三特异性 融合蛋白。In the previous studies of the inventors, it was found that the anti-CLD18A2 single-domain antibody had poor stability in vivo, and the application of IgG Fc, which is commonly used in the field to improve expression and stability, to be fused to it still cannot achieve substantial improvement. After extensive research and screening, the inventors fused the anti-CLD18A2 domain with the anti-CD3 domain and the anti-HSA domain to obtain an anti-CLD18A2/HSA/CD3 trispecific fusion protein.
如本文所用,所述的“抗CLD18A2/HSA/CD3三特异性融合蛋白”、“三特异性融合蛋白”、“三靶点靶向性的融合蛋白”可互换使用。As used herein, the "anti-CLD18A2/HSA/CD3 trispecific fusion protein", "trispecific fusion protein", and "triple target-targeting fusion protein" can be used interchangeably.
本发明的术语“特异性”表示本发明的三特异性融合蛋白不与或基本上不与或较少与目标抗原以外的其它蛋白交叉反应。其特异性的程度可以通过免疫学技术来判断,包括但不限于免疫印迹,免疫亲和层析,流式细胞分析等。例如,特异性识别优选通过流式细胞技术来确定,而具体情况下特异性识别的标准可由本领域一般技术人员根据其掌握的本领域常识来判断。The term "specificity" in the present invention means that the trispecific fusion protein of the present invention does not or substantially does not or less cross-reacts with other proteins other than the target antigen. The degree of its specificity can be judged by immunological techniques, including but not limited to western blotting, immunoaffinity chromatography, flow cytometry and the like. For example, the specific recognition is preferably determined by flow cytometry, and the criteria for specific recognition in specific cases can be judged by those of ordinary skill in the art based on their common knowledge in the field.
作为本发明的优选方式,所述的抗CLD18A2结构域为一种单域抗体,在本文中简称为“抗CLD18A2VHH”。所述的抗HSA结构域为一种单域抗体,在本文中简称为“抗HSA VHH”。本发明所提供的单域抗体,其整体分子量可以是ScFv单链抗体的二分之一左右,因此可以有效降低整体结构的分子量,从而增强其组织穿透性,更有效的到达靶组织和器官,提高治疗效果,且这种结构比起具有两个ScFv串联的结构更便于制备。所述的抗CD3结构域为一种单链抗体,在本发明中简称为“抗CD3scFv”。As a preferred mode of the present invention, the anti-CLD18A2 domain is a single domain antibody, referred to herein as "anti-CLD18A2VHH". The anti-HSA domain is a single domain antibody, which is referred to as "anti-HSA VHH" for short herein. The overall molecular weight of the single-domain antibody provided by the present invention can be about one-half that of the ScFv single-chain antibody, so the molecular weight of the overall structure can be effectively reduced, thereby enhancing its tissue penetration and reaching target tissues and organs more effectively , improve the therapeutic effect, and this structure is easier to prepare than the structure with two ScFvs in series. The anti-CD3 structural domain is a single-chain antibody, which is referred to as "anti-CD3 scFv" for short in the present invention.
作为本发明的优选方式,本发明人针对抗CLD18A2结构域和抗HSA结构域进行了优化改造,所述抗CLD18A2结构域的FR1区进行P→A突变;所述抗HSA结构域的FR1区进行P→A突变。所述突变非常显著地提高了融合蛋白的稳定性以及半衰期。As a preferred mode of the present invention, the inventors have optimized and transformed the anti-CLD18A2 domain and the anti-HSA domain, the FR1 region of the anti-CLD18A2 domain undergoes a P→A mutation; the FR1 region of the anti-HSA domain undergoes a mutation P→A mutation. The mutations very significantly increased the stability and half-life of the fusion protein.
作为本发明的优选方式,所述抗CD3结构域为单链抗体,较佳地其氨基酸序列如SEQ ID NO:1中第254~502位所示。As a preferred mode of the present invention, the anti-CD3 domain is a single-chain antibody, and preferably its amino acid sequence is shown in positions 254-502 of SEQ ID NO:1.
本发明中,所述抗CLD18A2结构域可特异性地结合肿瘤组织中的CLD18A2抗原;所述抗HSA结构域可以与血清中的白蛋白可逆地非共价结合;所述抗CD3scFv则可以与T细胞TCR复合物中的CD3亚基结合,激活T细胞,释放细胞因子,以杀伤肿瘤靶细胞。抗CLD18A2/HSA/CD3三特异性融合蛋白中各结构域的顺序(N端-C端)可以是:抗CLD18A2VHH-抗HSA VHH-抗CD3scFv或抗CLD18A2VHH-抗CD3scFv-抗HSA VHH。In the present invention, the anti-CLD18A2 domain can specifically bind to the CLD18A2 antigen in tumor tissue; the anti-HSA domain can reversibly and non-covalently bind to albumin in serum; the anti-CD3 scFv can bind to T The CD3 subunit in the cellular TCR complex binds, activates T cells, and releases cytokines to kill tumor target cells. The sequence (N-terminal-C-terminal) of each domain in the anti-CLD18A2/HSA/CD3 trispecific fusion protein can be: anti-CLD18A2VHH-anti-HSA VHH-anti-CD3scFv or anti-CLD18A2VHH-anti-CD3scFv-anti-HSA VHH.
在没有肿瘤细胞和T细胞存在的情况下,抗CLD18A2/HSA/CD3三特异性融合蛋白在外周血中与HSA非共价结合,通过HSA与FcRn结合的循环机制而延长其体内半衰期。在肿瘤微环境中,抗CLD18A2/HSA/CD3三特异性融合蛋白通过与表达CLD18A2抗原的肿瘤细胞和T细胞交联,产生TDCC效应(T-cell dependent cellular cytotoxicity)而杀伤肿瘤细胞。In the absence of tumor cells and T cells, the anti-CLD18A2/HSA/CD3 trispecific fusion protein non-covalently binds to HSA in peripheral blood, prolonging its in vivo half-life through the circulating mechanism of HSA binding to FcRn. In the tumor microenvironment, the anti-CLD18A2/HSA/CD3 trispecific fusion protein cross-links with tumor cells and T cells expressing CLD18A2 antigen to produce TDCC effect (T-cell dependent cellular cytotoxicity) to kill tumor cells.
所述融合蛋白中,还可以包括连接肽。例如,抗CLD18A2VHH和抗HSA  VHH之间、抗HSA VHH和抗CD3scFv之间可以设有连接肽。所述连接肽片段通常可以为一段长度合适的富含G、S和/或A(主要由甘氨酸(G)、丝氨酸(S)和/或丙氨酸(A)构成)的柔性多肽,从而使相邻的蛋白质结构域可相对于彼此自由移动。例如,所述连接肽片段的氨基酸序列可以包括如(GS)n、(GGS)n、(GGSG)n、(GGGS)nA、(GGGGS)nA、(GGGGS)nG、(GGGGA)nA、(GGGGG)nA等序列,其中,n选自1-10之间的整数。连接肽片段的氨基酸序列的长度可以为3-30、3-4、4-6、6-8、8-10、10-12、12-14、14-16、16-18、18-20、20-22、22-24、24-26、26-28或28-30。In the fusion protein, a connecting peptide may also be included. For example, anti-CLD18A2VHH and anti-HSA Linking peptides may be provided between VHHs, and between anti-HSA VHHs and anti-CD3 scFv. The connecting peptide fragment can generally be a flexible polypeptide rich in G, S and/or A (mainly composed of glycine (G), serine (S) and/or alanine (A)) with a suitable length, so that Adjacent protein domains are free to move relative to each other. For example, the amino acid sequence of the connecting peptide fragment may include (GS)n, (GGS)n, (GGSG)n, (GGGS)nA, (GGGGS)nA, (GGGGS)nG, (GGGGA)nA, (GGGGG )nA and other sequences, wherein n is selected from an integer between 1-10. The length of the amino acid sequence of the connecting peptide fragment can be 3-30, 3-4, 4-6, 6-8, 8-10, 10-12, 12-14, 14-16, 16-18, 18-20, 20-22, 22-24, 24-26, 26-28 or 28-30.
所述融合蛋白还可以包含标签蛋白,所述标签蛋白为6×His、Fc、Myc、GST、Flag或HA。在一些具体的实施方式中,所述标签蛋白为6×His,且所述标签蛋白的核苷酸序列如SEQ ID NO:9所示。The fusion protein may also contain a tag protein, and the tag protein is 6×His, Fc, Myc, GST, Flag or HA. In some specific embodiments, the tag protein is 6×His, and the nucleotide sequence of the tag protein is shown in SEQ ID NO:9.
在本发明中所列举的优选的结构域序列的基础上,本发明还包括它们的同功能变体。这些同功能变体与本发明所列举的优选的结构域的差别可以是氨基酸序列上的差异,也可以是不影响序列的修饰形式上的差异,或者兼而有之。应理解,所述变化更可能或通常发生在所述结构域的非CDR区,但是当考虑运用本发明优选的突变方案时,相应于抗CLD18A2结构域的FR1区P→A突变保持保守,相应于抗HSA结构域的FR1区P→A突变保持保守。On the basis of the preferred domain sequences listed in the present invention, the present invention also includes their homofunctional variants. The difference between these variants with the same function and the preferred structural domains listed in the present invention may be the difference in the amino acid sequence, or the difference in the modified form that does not affect the sequence, or both. It should be understood that the change is more likely or usually occurs in the non-CDR region of the domain, but when considering the preferred mutation scheme of the present invention, the P→A mutation in the FR1 region corresponding to the anti-CLD18A2 domain remains conservative, corresponding The P→A mutation in the FR1 region of the anti-HSA domain remains conservative.
在本发明中所列举的优选的结构域的基础上,本发明包括与所述的结构域序列同源性高(比如与所列举的具体蛋白序列的同源性为70%或更高;优选地同源性为80%或更高;更优选地同源性为90%或更高,如同源性95%,98%或99%)的、且具有相同功能的结构域。或者,本发明也包括经过一个或多个(如1-20个、1-15个、1-10个、1-8个、1-5个、1-3个或1-2个)氨基酸残基的取代、缺失或添加而形成的、且具有相同功能的结构域。同样地,所述变化更可能或通常发生在所述结构域的非CDR区,但是相应于SEQ ID NO:1所示的序列中第14位的Ala、第143位的Ala是保守的。On the basis of the preferred structural domains listed in the present invention, the present invention includes sequences with high homology with the described structural domains (such as 70% or higher homology with the specific protein sequences listed; preferably 80% or higher homology; more preferably 90% or higher homology, such as 95%, 98% or 99% homology), and have the same function. Alternatively, the present invention also includes one or more (such as 1-20, 1-15, 1-10, 1-8, 1-5, 1-3 or 1-2) amino acid residues A domain formed by substitution, deletion or addition of a group and having the same function. Likewise, the change is more likely or usually occurs in the non-CDR regions of the domain, but Ala at position 14, Ala at position 143 corresponding to the sequence shown in SEQ ID NO: 1 is conserved.
多核苷酸polynucleotide
本发明还提供了一种分离的多核苷酸,编码本发所述的抗CLD18A2/HSA/CD3三特异性融合蛋白,所述多核苷酸可以是RNA、DNA或cDNA等。提供所述分离的多核苷酸的方法对于本领域技术人员来说应该是已知的,例如,可以通过自动DNA合成和/或重组DNA技术等制备获得,也可以从适合的天然来源加以分离。在本发明一具体实施方式中,所述分离的多核苷酸的核酸序列可以包括如SEQ ID NO:4所示的序列。 The present invention also provides an isolated polynucleotide encoding the anti-CLD18A2/HSA/CD3 trispecific fusion protein of the present invention, the polynucleotide may be RNA, DNA or cDNA, etc. Methods for providing the isolated polynucleotide should be known to those skilled in the art, for example, it can be prepared by automatic DNA synthesis and/or recombinant DNA technology, etc., or it can be isolated from a suitable natural source. In a specific embodiment of the present invention, the nucleic acid sequence of the isolated polynucleotide may include the sequence shown in SEQ ID NO:4.
表达载体Expression vector
本发明还提供一种表达载体,所述表达载体含有本发明所述的分离的多核苷酸。所述表达载体的构建方法对于本领域技术人员来说应该是已知的,例如,所述表达载体可以通过体外重组DNA技术、DNA合成技术、体内重组技术等方法构建获得,更具体的,可以由所述的分离的多核苷酸插入到表达载体的多克隆位点构建而成。本发明中的表达载体通常指本领域熟知的各种市售表达载体等,例如可以是细菌质粒、噬菌体、酵母质粒、植物细胞病毒、哺乳动物细胞病毒如腺病毒、逆转录病毒或其他载体。所述载体还可以包括与所述多核苷酸序列操作性连接的一个或多个调控序列,所述调控序列可以包括合适的启动子序列。启动子序列通常与待表达氨基酸序列的编码序列操作性连接。启动子可以是在所选择的宿主细胞中显示转录活性的任何核苷酸序列,包括突变的、截短的和杂合启动子,并且可以从编码与该宿主细胞同源或异源的胞外或胞内多肽的基因获得。调控序列还可以包括合适的转录终止子序列,由宿主细胞识别以终止转录的序列。终止子序列与编码该多肽的核苷酸序列的3’末端相连,在选择的宿主细胞中有功能的任何终止子都可用于本发明。The present invention also provides an expression vector containing the isolated polynucleotide of the present invention. The method for constructing the expression vector should be known to those skilled in the art. For example, the expression vector can be constructed by methods such as in vitro recombinant DNA technology, DNA synthesis technology, and in vivo recombination technology. More specifically, it can be It is constructed by inserting the isolated polynucleotide into the multiple cloning site of the expression vector. The expression vector in the present invention generally refers to various commercially available expression vectors well known in the art, such as bacterial plasmids, phages, yeast plasmids, plant cell viruses, mammalian cell viruses such as adenovirus, retrovirus or other vectors. The vector may also include one or more regulatory sequences operably linked to the polynucleotide sequence, which may include a suitable promoter sequence. The promoter sequence is usually operably linked to the coding sequence for the amino acid sequence to be expressed. The promoter can be any nucleotide sequence that shows transcriptional activity in the host cell of choice, including mutated, truncated, and hybrid promoters, and can be derived from an extracellular sequence that encodes either homologous or heterologous to the host cell. Or intracellular polypeptide gene acquisition. The regulatory sequences may also include suitable transcription terminator sequences, sequences recognized by a host cell to terminate transcription. A terminator sequence is attached to the 3' end of the nucleotide sequence encoding the polypeptide, and any terminator that is functional in the host cell of choice may be used in the present invention.
通常来说,合适的载体可以包含在至少一种有机体中起作用的复制起点、启动子序列、方便的限制酶位点和一个或多个可选择的标记。例如,这些启动子可以是包括但不限于大肠杆菌的lac或trp启动子;λ噬菌体PL启动子;真核启动子包括CMV立即早期启动子、HSV胸苷激酶启动子、早期和晚期SV40启动子、毕赤酵母的甲醇氧化酶启动子和其它一些已知的可控制基因在原核或真核细胞或其病毒中表达的启动子。标记基因可用于提供用于选择转化的宿主细胞的表型性状,例如,可以是包括但不限于真核细胞培养用的二氢叶酸还原酶、新霉素抗性以及绿色荧光蛋白(GFP),或用于大肠杆菌的四环素或氨苄青霉素抗性等。当所述的多核苷酸被表达时,表达载体中还可以包括增强子序列,如果在载体中插入增强子序列,则将会使转录得到增强,增强子是DNA的顺式作用因子,通常大约有10到300个碱基对,作用于启动子以增强基因的转录。In general, suitable vectors will contain an origin of replication functional in at least one organism, a promoter sequence, convenient restriction enzyme sites and one or more selectable markers. For example, these promoters may be lac or trp promoters including but not limited to E. coli; bacteriophage lambda PL promoter; eukaryotic promoters including CMV immediate early promoter, HSV thymidine kinase promoter, early and late SV40 promoters , the methanol oxidase promoter of Pichia pastoris and some other known promoters that can control the expression of genes in prokaryotic or eukaryotic cells or their viruses. Marker genes may be used to provide phenotypic traits for selection of transformed host cells, for example, may include but are not limited to dihydrofolate reductase for eukaryotic cell culture, neomycin resistance, and green fluorescent protein (GFP), Or tetracycline or ampicillin resistance for E. coli etc. When the polynucleotide is expressed, an enhancer sequence can also be included in the expression vector. If the enhancer sequence is inserted into the vector, the transcription will be enhanced. The enhancer is a cis-acting factor of DNA, usually about There are 10 to 300 base pairs and act on the promoter to enhance the transcription of the gene.
表达系统expression system
本发明还提供一种抗CLD18A2/HSA/CD3三特异性融合蛋白的表达系统,所述表达系统含有本发明所述的表达载体或基因组中整合有外源的本发明所述的多核苷酸。任何适用于表达载体进行表达的细胞都可以作为宿主细胞,例如,所述宿主细胞可以是原核细胞,如细菌细胞;或是低等真核细胞,如酵母细胞;或是高等 真核细胞,如哺乳动物细胞,具体可以是包括但不限于大肠杆菌;链霉菌属;鼠伤寒沙门氏菌的细菌细胞;真菌细胞如酵母、丝状真菌、植物细胞;果蝇S2或Sf9的昆虫细胞;CHO、COS、HEK293细胞、或Bowes黑素瘤细胞的动物细胞等中的一种或多种的组合。构建所述表达系统的方法对于本领域技术人员来说应该是已知的,例如,可以是包括但不限于显微注射法、基因枪法、电穿孔法、病毒介导的转化法、电子轰击法、磷酸钙沉淀法等中的一种或多种的组合。作为本发明的优选方式,所述的表达系统为动物细胞表达系统,例如CHO,NS0,BHK,HEK-293或PER-C6等。在更具体的实施方式中,所述的细胞为CHO。The present invention also provides an expression system of an anti-CLD18A2/HSA/CD3 trispecific fusion protein, the expression system contains the expression vector of the present invention or the exogenous polynucleotide of the present invention is integrated in the genome. Any cell suitable for expressing the expression vector can be used as a host cell, for example, the host cell can be a prokaryotic cell, such as a bacterial cell; or a lower eukaryotic cell, such as a yeast cell; or a higher Eukaryotic cells, such as mammalian cells, specifically include but are not limited to Escherichia coli; Streptomyces sp; bacterial cells of Salmonella typhimurium; fungal cells such as yeast, filamentous fungi, plant cells; insect cells of Drosophila S2 or Sf9 ; A combination of one or more of CHO, COS, HEK293 cells, or animal cells such as Bowes melanoma cells. The method for constructing the expression system should be known to those skilled in the art, for example, it may include but not limited to microinjection method, particle gun method, electroporation method, virus-mediated transformation method, electron bombardment method , calcium phosphate precipitation method, etc. in one or more combinations. As a preferred mode of the present invention, the expression system is an animal cell expression system, such as CHO, NSO, BHK, HEK-293 or PER-C6. In a more specific embodiment, said cell is CHO.
融合蛋白的制备方法Preparation method of fusion protein
本发明还提供了所述抗CLD18A2/HSA/CD3三特异性融合蛋白的制备方法。本领域技术人员可选择合适的方法以制备所述融合蛋白,例如,所述制备方法可以包括:在适合表达所述融合蛋白的条件下,培养本发明所述的融合蛋白的表达系统,从而表达出所述的融合蛋白,纯化分离出所述的融合蛋白。The invention also provides a preparation method of the anti-CLD18A2/HSA/CD3 trispecific fusion protein. Those skilled in the art can choose an appropriate method to prepare the fusion protein. For example, the preparation method may include: cultivating the expression system of the fusion protein of the present invention under conditions suitable for expressing the fusion protein, so as to express The fusion protein is obtained, and the fusion protein is purified and isolated.
药物组合物/药盒Pharmaceutical composition/kit
本发明还提供一种药物组合物,包括本发明所述的抗CLD18A2/HSA/CD3三特异性融合蛋白、本发明所述的表达系统或其培养物。The present invention also provides a pharmaceutical composition, including the anti-CLD18A2/HSA/CD3 trispecific fusion protein of the present invention, the expression system of the present invention or its culture.
所述药物组合物中还可以包括各种本领域药学上可接受的载体。所述药学上可接受的载体可以包括各种赋形剂和稀释剂,这些载体本身并不是必要的活性成分,且施用后没有过分的毒性。合适的载体对于本领域技术人员来说应该是熟知的,例如,在Remington’s Pharmaceutical Sciences(Mack Pub.Co.,N.J.,1991)中可找到关于药学上可接受的载体的充分讨论。Various pharmaceutically acceptable carriers in the art may also be included in the pharmaceutical composition. The pharmaceutically acceptable carrier may include various excipients and diluents, these carriers themselves are not essential active ingredients, and there is no excessive toxicity after administration. Suitable carriers will be well known to those skilled in the art, for example, a full discussion of pharmaceutically acceptable carriers can be found in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J., 1991).
所述药物组合物中,融合蛋白含量通常是有效量的,该有效量所对应的活性成分的含量可以根据所治疗的对象和特定给药方式来确定。In the pharmaceutical composition, the content of the fusion protein is usually an effective amount, and the content of the active ingredient corresponding to the effective amount can be determined according to the object to be treated and the specific administration method.
本发明也提供了含有所述的融合蛋白、所述的表达系统或其表达产物(培养物)或所述的药物组合物的药盒。所述的药盒中还可包括使用说明书,以指导本领域技术人员或临床医师的应用。The present invention also provides a kit containing the fusion protein, the expression system or its expression product (culture) or the pharmaceutical composition. Instructions for use can also be included in the kit to guide the application of those skilled in the art or clinicians.
用途use
本发明还提供了所述的抗CLD18A2/HSA/CD3三特异性融合蛋白、所述的表达系统或其培养物在制备用于缓解或治疗CLD18A2阳性细胞相关疾病的药物中的用途。 The present invention also provides the use of the anti-CLD18A2/HSA/CD3 trispecific fusion protein, the expression system or its culture in the preparation of medicines for alleviating or treating diseases related to CLD18A2 positive cells.
如本文所用,所述的“CLD18A2阳性细胞(相关)疾病”包括“CLD18A2异常表达(相关)疾病”或“CLD18A2高表达(相关)疾病”,在未特意进行区别性描述的情况下这些术语可互换使用。As used herein, the "CLD18A2-positive cell (related) disease" includes "CLD18A2 abnormal expression (related) disease" or "CLD18A2 high expression (related) disease", and these terms may be Used interchangeably.
CLD18A2在很多肿瘤细胞中表达。鉴于CLD18A2的这种特性,CLD18A2可以作为相关肿瘤的治疗靶点。本发明中,所述CLD18A2阳性细胞可以包括所有表达CLD18A2的肿瘤,这些肿瘤通常可以为CLD18A2阳性表达。CLD18A2的阳性表达通常指出现CLD18A2的表达、或CLD18A2的表达水平高于一定的标准,例如,CLD18A2阳性可以是肿瘤组织中可检测出CLD18A2的mRNA的表达,再例如,CLD18A2阳性可以是肿瘤组织中可检测出CLD18A2的蛋白的表达,再例如,CLD18A2阳性可以是肿瘤组织的CLD18A2的mRNA表达水平高于其周围健康组织,再例如,CLD18A2阳性可以是肿瘤组织的CLD18A2蛋白的表达水平高于其周围健康组织。这些疾病具体可以包括但不限于胃癌、食管癌、胰腺癌、肺癌、卵巢癌、乳腺癌、结肠直肠癌、肝癌、胆囊癌和头颈癌等,这些癌症可以为早期、中期或晚期等,例如转移癌等。CLD18A2 is expressed in many tumor cells. In view of this characteristic of CLD18A2, CLD18A2 can be used as a therapeutic target for related tumors. In the present invention, the CLD18A2-positive cells may include all CLD18A2-expressing tumors, and these tumors may usually express CLD18A2-positively. The positive expression of CLD18A2 usually refers to the expression of CLD18A2, or the expression level of CLD18A2 is higher than a certain standard. The expression of CLD18A2 protein can be detected. For another example, CLD18A2 positive can be that the mRNA expression level of CLD18A2 in the tumor tissue is higher than that of its surrounding healthy tissue. For another example, CLD18A2 positive can be that the expression level of CLD18A2 protein in the tumor tissue is higher than its surrounding healthy tissue. These diseases may specifically include but are not limited to gastric cancer, esophageal cancer, pancreatic cancer, lung cancer, ovarian cancer, breast cancer, colorectal cancer, liver cancer, gallbladder cancer, head and neck cancer, etc. These cancers may be early, middle or late, such as metastasis cancer etc.
治疗方法treatment method
本发明还提供一种治疗方法包括:向个体施用有效量的所述的抗CLD18A2/HSA/CD3三特异性融合蛋白、所述的表达系统或其培养物、或所述的药物组合物。The present invention also provides a treatment method comprising: administering to an individual an effective amount of the anti-CLD18A2/HSA/CD3 trispecific fusion protein, the expression system or its culture, or the pharmaceutical composition.
本发明所提供的融合蛋白、药物组合物的“治疗有效量”优选地导致疾病症状的严重性降低,疾病无症状期的频率和持续时间增加,或者防止因疾病痛苦而引起的损伤或失能。例如,对于CLD18A2相关肿瘤的治疗(包括如胃癌),相对于未接受治疗的对象,“治疗有效量”优选地将细胞生长或肿瘤生长抑制至少约10%,优选至少约20%,更优选至少约30%,更优选至少约40%,更优选至少约50%,更优选至少约60%,更优选至少约70%,更优选至少约80%。抑制肿瘤生长的能力可以在预测对人类肿瘤的疗效的动物模型系统中评价。或者,也可以通过检查抑制细胞生长的能力来评价,这种抑制可以通过本领域技术人员公知的试验在体外测定。治疗有效量的融合蛋白、药物组合物通常能够减小肿瘤大小,或者以其他方式缓解对象的症状。本领域技术人员可以根据实际情况选择合适的治疗有效量,例如,可以是对象的大小、对象症状的严重性和选择的特定组合物或给药途径。治疗的处方(例如,对剂量的决定等)可以是由医生确定的,通常考虑的因素包括但不限于所治疗的疾病、患者个体的情况、递送部位、施用方法以及其它因素等。预防有效量指在必需的剂量和时间上有效实现期望的预防效果的 量。The "therapeutically effective amount" of the fusion protein and pharmaceutical composition provided by the present invention preferably leads to a reduction in the severity of disease symptoms, an increase in the frequency and duration of the asymptomatic period of the disease, or prevention of damage or disability caused by the suffering of the disease . For example, for the treatment of CLD18A2-related tumors (including, for example, gastric cancer), a "therapeutically effective amount" preferably inhibits cell growth or tumor growth by at least about 10%, preferably at least about 20%, more preferably at least About 30%, more preferably at least about 40%, more preferably at least about 50%, more preferably at least about 60%, more preferably at least about 70%, more preferably at least about 80%. The ability to inhibit tumor growth can be evaluated in animal model systems predictive of efficacy against human tumors. Alternatively, it can also be assessed by examining the ability to inhibit cell growth, which can be determined in vitro by assays well known to those skilled in the art. A therapeutically effective amount of a fusion protein, pharmaceutical composition, generally reduces tumor size, or otherwise alleviates symptoms in a subject. Those skilled in the art can select an appropriate therapeutically effective amount according to the actual situation, for example, the size of the subject, the severity of the subject's symptoms and the selected specific composition or administration route. Prescriptions for treatment (e.g., dosage decisions, etc.) can be determined by a physician, generally taking into account factors including, but not limited to, the disease being treated, individual patient conditions, site of delivery, method of administration, and other factors. Prophylactically effective dose refers to the amount that is effective in achieving the desired preventive effect at the necessary dose and time. quantity.
本发明的融合蛋白和药物组合物可以作为单一有效成分施用,也可以在联合治疗中施用,即与其他药剂联用。例如,所述联合治疗可以是所述融合蛋白、药物组合物联合其他至少一种抗肿瘤药物(例如,紫杉醇等)。再例如,所述联合治疗可以是所述融合蛋白、药物组合物与免疫检查点抑制剂联合使用,所述免疫检查点抑制剂包括但不限于PD-1抑制剂、PD-L1抑制剂、或CTLA-4抑制剂等中的一种或多种的组合,所述抑制剂可以是单克隆抗体。The fusion protein and pharmaceutical composition of the present invention can be administered as a single active ingredient, or in combination therapy, that is, in combination with other agents. For example, the combination therapy may be the fusion protein or the pharmaceutical composition combined with at least one other anti-tumor drug (for example, paclitaxel, etc.). For another example, the combination therapy may be the combination of the fusion protein, the pharmaceutical composition and an immune checkpoint inhibitor, and the immune checkpoint inhibitor includes but is not limited to a PD-1 inhibitor, a PD-L1 inhibitor, or A combination of one or more of CTLA-4 inhibitors, etc., said inhibitors may be monoclonal antibodies.
以下通过特定的具体实例说明本发明的实施方式,本领域技术人员可由本说明书所揭露的内容轻易地了解本发明的其他优点与功效。本发明还可以通过另外不同的具体实施方式加以实施或应用,本说明书中的各项细节也可以基于不同观点与应用,在没有背离本发明的精神下进行各种修饰或改变。Embodiments of the present invention are described below through specific examples, and those skilled in the art can easily understand other advantages and effects of the present invention from the content disclosed in this specification. The present invention can also be implemented or applied through other different specific implementation modes, and various modifications or changes can be made to the details in this specification based on different viewpoints and applications without departing from the spirit of the present invention.
在进一步描述本发明具体实施方式之前,应理解,本发明的保护范围不局限于下述特定的具体实施方案;还应当理解,本发明实施例中使用的术语是为了描述特定的具体实施方案,而不是为了限制本发明的保护范围。Before further describing the specific embodiments of the present invention, it should be understood that the protection scope of the present invention is not limited to the following specific specific embodiments; it should also be understood that the terms used in the examples of the present invention are to describe specific specific embodiments, It is not intended to limit the protection scope of the present invention.
当实施例给出数值范围时,应理解,除非本发明另有说明,每个数值范围的两个端点以及两个端点之间任何一个数值均可选用。除非另外定义,本发明中使用的所有技术和科学术语与本技术领域技术人员通常理解的意义相同。除实施例中使用的具体方法、设备、材料外,根据本技术领域的技术人员对现有技术的掌握及本发明的记载,还可以使用与本发明实施例中所述的方法、设备、材料相似或等同的现有技术的任何方法、设备和材料来实现本发明。When the examples give numerical ranges, it should be understood that, unless otherwise stated in the present invention, the two endpoints of each numerical range and any value between the two endpoints can be selected. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition to the specific methods, equipment, and materials used in the examples, according to those skilled in the art's grasp of the prior art and the description of the present invention, the methods, equipment, and materials described in the examples of the present invention can also be used Any methods, apparatus and materials of the prior art similar or equivalent to the practice of the present invention.
除非另外说明,本发明中所公开的实验方法、检测方法、制备方法均采用本技术领域常规的分子生物学、生物化学、染色质结构和分析、分析化学、细胞培养、重组DNA技术及相关领域的常规技术。这些技术在现有文献中已有完善说明,具体可参见Sambrook等MOLECULAR CLONING:A LABORATORY MANUAL,Second edition,Cold Spring Harbor Laboratory Press,1989and Third edition,2001;Ausubel等,CURRENT PROTOCOLS IN MOLECULAR BIOLOGY,John Wiley&Sons,New York,1987and periodic updates;the series METHODS IN ENZYMOLOGY,Academic Press,San Diego;Wolffe,CHROMATIN STRUCTURE AND FUNCTION,Third edition,Academic Press,San Diego,1998;METHODS IN ENZYMOLOGY,Vol.304,Chromatin(P.M.Wassarman and A.P.Wolffe,eds.),Academic Press,San Diego,1999;和METHODS IN MOLECULAR BIOLOGY,Vol.119,Chromatin  Protocols(P.B.Becker,ed.)Humana Press,Totowa,1999等。Unless otherwise stated, the experimental methods, detection methods, and preparation methods disclosed in the present invention all adopt conventional molecular biology, biochemistry, chromatin structure and analysis, analytical chemistry, cell culture, recombinant DNA technology and related fields in the technical field conventional technology. These techniques have been fully described in the existing literature. For details, see Sambrook et al. MOLECULAR CLONING: A LABORATORY MANUAL, Second edition, Cold Spring Harbor Laboratory Press, 1989 and Third edition, 2001; Ausubel et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons , New York, 1987 and periodic updates; the series METHODS IN ENZYMOLOGY, Academic Press, San Diego; Wolffe, CHROMATIN STRUCTURE AND FUNCTION, Third edition, Academic Press, San Diego, 1998; METHODS IN ENZYMOLOGY, Vol.304, Ch romatin (PM Wassarman and AP Wolffe, eds.), Academic Press, San Diego, 1999; and METHODS IN MOLECULAR BIOLOGY, Vol.119, Chromatin Protocols (PB Becker, ed.) Humana Press, Totowa, 1999 et al.
实施例1、三特异性融合蛋白的融合表达Embodiment 1, the fusion expression of trispecific fusion protein
1、三特异性融合蛋白的设计1. Design of trispecific fusion protein
DR303X8序列(SEQ ID NO:1):
DR303X8 sequence (SEQ ID NO: 1):
上述序列中,第14位和第143位的“A”为发明人引入的P/A突变位点。第1~120位为抗CLDN18.2VHH,第130~244位为抗HSA VHH,第254~502位为抗CD3scFv。In the above sequence, the "A" at the 14th and 143rd positions is the P/A mutation site introduced by the inventors. Positions 1-120 are anti-CLDN18.2 VHH, positions 130-244 are anti-HSA VHH, positions 254-502 are anti-CD3 scFv.
斜体下划线标示的氨基酸为连接序列。Amino acids underlined in italics are linker sequences.
下划线加粗标示的氨基酸片段为CDR。其中,对于抗CLDN18.2VHH,第26~33位为CDR1,第51~57位为CDR2,第96~109位为CDR3。对于抗HSA VHH,第155~162位为CDR1,第178~187位为CDR2,第226~233位为CDR3。对于抗CD3scFv,第279~286位为HCDR1,第304~313位为HCDR2,第352~367位为HCDR3;第419~428位为LCDR1,第445~447位为LCDR2,第484~492位为LCDR3。采用IMGT编号方案(antibody numbering scheme)进行CDR区标注。Amino acid fragments marked in bold underline are CDRs. Among them, for the anti-CLDN18.2VHH, the 26th to 33rd positions are CDR1, the 51st to 57th positions are CDR2, and the 96th to 109th positions are CDR3. For anti-HSA VHH, positions 155-162 are CDR1, positions 178-187 are CDR2, and positions 226-233 are CDR3. For anti-CD3 scFv, the 279th-286th position is HCDR1, the 304th-313rd position is HCDR2, the 352th-367th position is HCDR3; the 419th-428th position is LCDR1, the 445th-447th position is LCDR2, and the 484th-492nd position is LCDR3. The CDR region was marked using the IMGT numbering scheme (antibody numbering scheme).
DR303X6序列(SEQ ID NO:2):
DR303X6 sequence (SEQ ID NO:2):
上述序列中,第1~120位为抗CLDN18.2VHH;第130~378位为抗CD3scFv(不含抗HSA VHH)。In the above sequence, positions 1-120 are anti-CLDN18.2 VHH; positions 130-378 are anti-CD3 scFv (excluding anti-HSA VHH).
下划线加粗标示的氨基酸片段为CDR。其中,对于抗CLDN18.2VHH,第26~33位为CDR1,第51~57位为CDR2,第96~109位为CDR3。对于抗CD3scFv,第155~162为HCDR1,第180~189位为HCDR2,第228~243位为HCDR3;第 295~303位为LCDR1,第321~323位为LCDR2,第360~368位为LCDR3。采用IMGT编号方案(antibody numbering scheme)进行CDR区标注。Amino acid fragments marked in bold underline are CDRs. Among them, for the anti-CLDN18.2VHH, the 26th to 33rd positions are CDR1, the 51st to 57th positions are CDR2, and the 96th to 109th positions are CDR3. For anti-CD3 scFv, 155th to 162th is HCDR1, 180th to 189th is HCDR2, 228th to 243rd is HCDR3; Bits 295 to 303 are LCDR1, bits 321 to 323 are LCDR2, and bits 360 to 368 are LCDR3. The CDR region was marked using the IMGT numbering scheme (antibody numbering scheme).
DR303X2序列(SEQ ID NO:3)
DR303X2 sequence (SEQ ID NO: 3)
上述序列中,第1~120位为抗CLDN18.2VHH,第130~244位为抗HSA VHH,第254~502位为抗CD3scFv。In the above sequence, positions 1-120 are anti-CLDN18.2 VHH, positions 130-244 are anti-HSA VHH, positions 254-502 are anti-CD3 scFv.
斜体下划线标示的氨基酸为连接序列。Amino acids underlined in italics are linker sequences.
下划线加粗标示的氨基酸片段为CDR。其中,对于抗CLDN18.2VHH,第26~33位为CDR1,第51~57位为CDR2,第96~109位为CDR3。对于抗HSA VHH,第155~162位为CDR1,第178~187位为CDR2,第226~233位为CDR3。对于抗CD3scFv,第279~286位为HCDR1,第304~313位为HCDR2,第352~367位为HCDR3;第419~428位为LCDR1,第445~447位为LCDR2,第484~492位为LCDR3。采用IMGT编号方案(antibody numbering scheme)进行CDR区标注。Amino acid fragments marked in bold underline are CDRs. Among them, for the anti-CLDN18.2VHH, the 26th to 33rd positions are CDR1, the 51st to 57th positions are CDR2, and the 96th to 109th positions are CDR3. For anti-HSA VHH, positions 155-162 are CDR1, positions 178-187 are CDR2, and positions 226-233 are CDR3. For anti-CD3 scFv, the 279th-286th position is HCDR1, the 304th-313rd position is HCDR2, the 352th-367th position is HCDR3; the 419th-428th position is LCDR1, the 445th-447th position is LCDR2, and the 484th-492nd position is LCDR3. The CDR region was marked using the IMGT numbering scheme (antibody numbering scheme).
2、重组表达2. Recombinant expression
将抗CLD18A2结构域、抗CD3结构域和抗人血清白蛋白(HSA)结构域,通过连接肽串联组合成抗CLD18A2/HSA/CD3三特异性融合蛋白(表1),并委托通用生物系统(安徽)有限公司进行碱基序列的优化合成,在5’端添加信号肽(METDTLLLWVLLLWVPGSTG)后,构建至动物细胞表达载体进行分泌表达。用去内毒素质粒大抽试剂盒(Biomiga)提取重组表达质粒,将质粒和转染试剂以质量比1:3的比例量取,并分别用培养基稀释,将稀释液混合后室温静置30min后加入到CHO细胞中,37℃,5%CO2摇床培养箱中培养7天后,离心获取细胞表达上清,用于纯化。The anti-CLD18A2 domain, anti-CD3 domain and anti-human serum albumin (HSA) domain were combined in tandem through connecting peptides to form an anti-CLD18A2/HSA/CD3 trispecific fusion protein (Table 1), and entrusted to General Biosystems ( Anhui) Co., Ltd. optimized the synthesis of the base sequence. After adding a signal peptide (METDTLLLWVLLLWVPGSTG) at the 5' end, it was constructed into an animal cell expression vector for secreted expression. Use the endotoxin-free plasmid extraction kit (Biomiga) to extract the recombinant expression plasmid, measure the plasmid and transfection reagent at a mass ratio of 1:3, and dilute them with medium respectively, mix the dilutions and let stand at room temperature for 30 minutes After that, it was added to CHO cells, cultured in a shaker incubator at 37° C. with 5% CO 2 for 7 days, and centrifuged to obtain cell expression supernatant for purification.
表1

Table 1

X910:根据US_2020_0055932_A1的SEQ ID NO:132序列,在CHO细胞中表达制备。X910: According to the sequence of SEQ ID NO: 132 of US_2020_0055932_A1, it was expressed and prepared in CHO cells.
3、纯化制备3. Purification and preparation
细胞表达上清液高速离心,然后经1.2μm膜过滤后,上样至AT Protein A Diamond亲和层析柱(博格隆(上海)生物技术有限公司)。平衡液为20mmol/L Tris-HCl,150mmol/L NaCl,pH7.4溶液;淋洗液为20mmol/L HAc-NaAc pH5.0溶液;洗脱液为100mmol/L Gly-HCl,pH3.0溶液;清洗液为0.1mmol/L NaOH溶液;中和液为1mol/L Tris-HCl,pH8.5。用平衡液平衡层析柱,以5min保留时间将样品按载量不高于20mg目标蛋白/ml填料上样,上样后用平衡液平衡层析柱,再以淋洗液淋洗3-5倍柱床体积,最后用洗脱液洗脱目标峰,将目标峰用中和液调pH至7.0,离心去除沉淀,取上清。将离心上清调节电导至低于5mS/cm,过1.2μm滤膜后上样至Q Bestarose FF阴离子交换层析柱(博格隆(上海)生物技术有限公司),流穿模式去除杂质,平衡液为20mmol/L PB,pH7.0溶液。将阴离子交换流穿样品上样至Diamond SP阳离子交换层析柱(博格隆(上海)生物技术有限公司),0-100%B线性梯度洗脱(0-500mmol/L NaCl,20mmol/L PB,pH7.0溶液,20CV),获得SDS-PAGE电泳纯度≥95%样品。The cell expression supernatant was centrifuged at high speed, then filtered through a 1.2 μm membrane, and loaded onto an AT Protein A Diamond affinity chromatography column (Borgeron (Shanghai) Biotechnology Co., Ltd.). Equilibrium solution is 20mmol/L Tris-HCl, 150mmol/L NaCl, pH7.4 solution; eluent is 20mmol/L HAc-NaAc pH5.0 solution; eluent is 100mmol/L Gly-HCl, pH3.0 solution ; The cleaning solution is 0.1mmol/L NaOH solution; the neutralizing solution is 1mol/L Tris-HCl, pH8.5. Equilibrate the chromatographic column with the balance liquid, load the sample with a retention time of 5 minutes according to the load of no more than 20 mg target protein/ml filler, equilibrate the chromatographic column with the balance liquid after loading the sample, and then rinse with the eluent for 3-5 times the volume of the column bed, and finally elute the target peak with the eluent, adjust the pH of the target peak to 7.0 with neutralizing solution, centrifuge to remove the precipitate, and take the supernatant. Adjust the conductance of the centrifuged supernatant to less than 5mS/cm, pass through a 1.2μm filter membrane, and then load the sample onto the Q Bestarose FF anion exchange chromatography column (Borgeron (Shanghai) Biotechnology Co., Ltd.), the flow-through mode removes impurities, and balances The solution is 20mmol/L PB, pH7.0 solution. The anion-exchange flow-through sample was loaded onto a Diamond SP cation-exchange chromatography column (Borgeron (Shanghai) Biotechnology Co., Ltd.), 0-100% B linear gradient elution (0-500mmol/L NaCl, 20mmol/L PB , pH7.0 solution, 20CV), obtain SDS-PAGE electrophoresis purity≥95% sample.
表达结果显示,即使带有改善表达量和稳定性的IgG Fc,X910与DR303X8相比,表达量偏低,分别为0.67g/L和2.27g/L。The expression results showed that even with IgG Fc with improved expression and stability, the expression of X910 was lower than that of DR303X8, which were 0.67g/L and 2.27g/L, respectively.
实施例2、体外活性检测Embodiment 2, in vitro activity detection
CHO-C18.2-gfpx稳转细胞株(高表达CLDN18A2的靶细胞)的构建:将含有CLD18A2全长基因(序列如SEQ ID NO:7所示)和egfp基因(序列如SEQ ID NO:8所示)的双表达框质粒pDR05-C18.2-gfp,用无内毒素质粒试剂盒提取质粒,无菌过滤后电转CHO-S细胞,并进行96孔板铺板添加MSX加压培养。挑取96孔板上的呈单独成团生长的稳转细胞,在添加MSX的培养条件下,逐步放大,并用制备的阳性对照抗体zolbetuximab类似物(专利US9751934B2中的序列号118的重链和序列号125的轻链)进行免疫荧光检测以及用Anti-Claudin18抗体[34H14L15](Abcam)进行斑 点印迹杂交(Dot blotting)检测,获得CLD18A2阳性表达细胞株CHO-C18.2-gfpx.稳转细胞株。Construction of CHO-C18.2-gfpx stably transfected cell line (target cell highly expressing CLDN18A2): CLD18A2 full-length gene (sequence shown in SEQ ID NO: 7) and egfp gene (sequence shown in SEQ ID NO: 8 Shown) double expression cassette plasmid pDR05-C18.2-gfp, the plasmid was extracted with an endotoxin-free plasmid kit, electroporated into CHO-S cells after sterile filtration, and cultured in 96-well plates with MSX under pressure. Pick the stably transfected cells growing in separate clusters on the 96-well plate, and gradually amplify them under the culture condition of adding MSX, and use the prepared positive control antibody zolbetuximab analog (the heavy chain and sequence number of sequence number 118 in the patent US9751934B2 No. 125 light chain) for immunofluorescence detection and Anti-Claudin18 antibody [34H14L15] (Abcam) for spotting The CLD18A2 positive expression cell line CHO-C18.2-gfpx. stably transfected cell line was obtained by Dot blotting detection.
Jurkat-NFAT-Luc2p(效应细胞)稳转细胞株的构建:Jurkat细胞转入NFAT响应的荧光素酶系统,获得TDCC活性检测效应细胞株Jurkat-NFAT-Luc2p。Construction of Jurkat-NFAT-Luc2p (effector cell) stably transfected cell line: Jurkat cells were transformed into the NFAT-responsive luciferase system to obtain the TDCC activity detection effector cell line Jurkat-NFAT-Luc2p.
体外活性检测采用T细胞激活荧光信号通路法。分别取适量CHO-C18.2-gfpx与Jurkat-NFAT-Luc2p细胞,1000rpm离心5/10min,弃去上清,加入RPMI1640培养基(含1%FBS)重悬,计数稀释至指定浓度,按一定比例(E:T=1:4,T=15000)两种细胞混匀加入到96孔细胞培养板中(50μl/孔),备用。用RPMI1640培养基(含1%FBS)稀释蛋白至指定浓度,然后取适量体积(25μl/孔)加入到96孔板内,过夜培养18个小时。加入检测溶液(荧光素酶检测试剂盒,Promega,E2620)10μl/孔,震荡1.5min,转移溶液60μl至白板中,于酶标仪(Molecular Devices I3)中检测。In vitro activity detection adopts T cell activation fluorescence signaling pathway method. Take an appropriate amount of CHO-C18.2-gfpx and Jurkat-NFAT-Luc2p cells, centrifuge at 1000rpm for 5/10min, discard the supernatant, add RPMI1640 medium (containing 1% FBS) to resuspend, count and dilute to the specified concentration, press a certain The ratio (E:T=1:4, T=15000) of the two kinds of cells was mixed and added to a 96-well cell culture plate (50 μl/well) for later use. The protein was diluted to the specified concentration with RPMI1640 medium (containing 1% FBS), and then an appropriate volume (25 μl/well) was added to a 96-well plate, and cultured overnight for 18 hours. Add 10 μl/well of detection solution (luciferase detection kit, Promega, E2620), shake for 1.5 min, transfer 60 μl of the solution to a white plate, and detect in a microplate reader (Molecular Devices I3).
结果显示,DR303X6、DR303X8、X910的EC50值分别约为69、61和75pM。抗HSA VHH的存在并不影响融合蛋白的体外活性。在另一次独立的活性检测实验中,DR303X2的EC50值约为75pM。The results showed that the EC 50 values of DR303X6, DR303X8 and X910 were about 69, 61 and 75pM, respectively. The presence of anti-HSA VHH did not affect the in vitro activity of the fusion protein. In another independent activity assay, the EC 50 value of DR303X2 was about 75pM.
实施例3、血清稳定性Embodiment 3, serum stability
采用实施例2中的报告基因法来比较DR303X2和DR303X8在37℃下、人血清中孵育7天的稳定性。The reporter gene method in Example 2 was used to compare the stability of DR303X2 and DR303X8 incubated in human serum for 7 days at 37°C.
取适量CHO-C18.2-gfpx与Jurkat-NFAT-Luc2p-10F2细胞,1000rpm离心5/10min,弃去上清,加入1640培养基(含1%FBS)重悬,计数,使用分析培养基调整效应细胞Jurkat-NFAT-luc2p-10F2细胞密度至2.4×106cells/ml,调整靶细胞CHO-C18.2-gfpx细胞密度至6×105cells/ml,1:1混合后铺96孔板,每孔50μl,备用。用1640培养基(含1%FBS)稀释蛋白至指定浓度(起始浓度为2μg/ml,配制浓度×2,5倍梯度稀释),然后取50μl加入到96孔板内,孵育培养18个小时。加入检测溶液(荧光素酶检测试剂盒,Promega,E2620)20μl/孔,震荡1min 30s,于酶标仪中检测光度值。平行检测2个样品的TDCC活性。Take an appropriate amount of CHO-C18.2-gfpx and Jurkat-NFAT-Luc2p-10F2 cells, centrifuge at 1000rpm for 5/10min, discard the supernatant, add 1640 medium (containing 1% FBS) to resuspend, count, and use the analysis medium to adjust Effector cell Jurkat-NFAT-luc2p-10F2 cell density to 2.4×10 6 cells/ml, adjust target cell CHO-C18.2-gfpx cell density to 6×10 5 cells/ml, mix 1:1 and spread in 96-well plate , 50 μl per well, spare. Dilute the protein with 1640 medium (containing 1% FBS) to the specified concentration (initial concentration is 2μg/ml, prepared concentration × 2, 5-fold gradient dilution), then take 50μl and add it to a 96-well plate, and incubate for 18 hours . Add 20 μl/well of detection solution (luciferase detection kit, Promega, E2620), shake for 1 min 30 s, and detect the photometric value in a microplate reader. The TDCC activity of the two samples was detected in parallel.
DR303X2样品和DR303X8样品在人血清中随着37℃放置时间的递增,TDCC活性都呈现下降的趋势。The TDCC activity of DR303X2 and DR303X8 samples in human serum showed a downward trend with increasing storage time at 37°C.
其中,DR303X2样品在人血清(HS)中0天的EC50为0.04012,7天的EC50为0.07349,活性残留率为55%。Among them, the EC 50 of the DR303X2 sample in human serum (HS) was 0.04012 on day 0, and the EC 50 on day 7 was 0.07349, and the residual activity rate was 55%.
DR303X8样品在在人血清(HS)中0天的EC50为0.01426,7天的EC50为0.02265,活性残留率为63%。The EC 50 of the DR303X8 sample in human serum (HS) was 0.01426 at day 0, and the EC 50 at day 7 was 0.02265, and the residual activity rate was 63%.
活性残留率=EC50(0天)/EC50(7天)*100%。 Activity residual rate=EC 50 (0 day) /EC 50 (7 day) *100%.
因此,DR303X8相较于DR303X2在人血清中具有更好的稳定性。Therefore, DR303X8 has better stability in human serum than DR303X2.
实施例4、C57BL/6小鼠体内半衰期Embodiment 4, half-life in vivo of C57BL/6 mice
C57BL/6小鼠尾静脉注射单次给予1mg/kg DR303X2和1mg/kg DR303X8,采集药前3天、药后1h、6h、24h、30h、48h、72h、96h、120h血样分析药物浓度。如图1所示,并使用PK solver 2.0软件采用非房室模型计算药代参数。DR303X2和DR303X8小鼠尾静脉给药剂量为1mg/kg时,半衰期分别为26.8h和28.8h。C57BL/6 mice were given 1 mg/kg DR303X2 and 1 mg/kg DR303X8 by tail vein injection, and blood samples were collected 3 days before the drug, 1 h, 6 h, 24 h, 30 h, 48 h, 72 h, 96 h, and 120 h after the drug to analyze the drug concentration. As shown in Figure 1, the pharmacokinetic parameters were calculated using the non-compartmental model using PK solver 2.0 software. When the dose of DR303X2 and DR303X8 mice was administered through the tail vein of 1mg/kg, the half-lives were 26.8h and 28.8h respectively.
实施例5、食蟹猴体内半衰期Embodiment 5, half-life in cynomolgus monkeys
食蟹猴单次静脉分别给予0.03mg/kg、0.6mg/kg、3mg/kg DR303X8或DR303X6,给药前、给药后即刻、4h、8h、24h、48h、72h、120h和168h采样分析血药浓度,并使用非房室模型计算药代参数。Cynomolgus monkeys were given 0.03mg/kg, 0.6mg/kg, 3mg/kg DR303X8 or DR303X6 intravenously in a single dose, and the blood was sampled and analyzed before administration, immediately after administration, 4h, 8h, 24h, 48h, 72h, 120h and 168h Drug concentrations were calculated, and pharmacokinetic parameters were calculated using non-compartmental models.
DR303X8或DR303X6静脉注射至食蟹猴体内:给药剂量0.03mg/kg时,Cmax为1.02±0.02μg/ml,AUC0-t为33.12±0.12μg/ml*h,t1/2为46.03±16.32h;给药剂量0.6mg/kg时,Cmax为17.33±0.65μg/ml,AUC0-t为641.74±102.66μg/ml*h,t1/2为37.37±1.11h;给药剂量3mg/kg时Cmax为81.01±2.44μg/ml,AUC0-t为3846.04±497.80μg/ml*h,t1/2为41.8±5.76h。DR303X8 or DR303X6 was intravenously injected into cynomolgus monkeys: when the dose was 0.03mg/kg, the C max was 1.02±0.02μg/ml, the AUC 0-t was 33.12±0.12μg/ml*h, and the t 1/2 was 46.03 ±16.32h; when the dose was 0.6mg/kg, C max was 17.33±0.65μg/ml, AUC 0-t was 641.74±102.66μg/ml*h, t 1/2 was 37.37±1.11h; At 3mg/kg, C max was 81.01±2.44μg/ml, AUC 0-t was 3846.04±497.80μg/ml*h, t 1/2 was 41.8±5.76h.
DR303X8静脉注射,在高中低剂量组给药浓度(3mg/kg、0.6mg/kg、0.03mg/kg)比为100:20:1时,Cmax比为80:17:1,AUC0-t比为116:20:1,系统暴露量与给药剂量线性相关。高中低剂量组半衰期、保留时间、清除率均没有明显差异,如图2。Intravenous injection of DR303X8, when the administration concentration (3mg/kg, 0.6mg/kg, 0.03mg/kg) ratio of 100:20:1 in the high, middle and low dose groups, the C max ratio was 80:17:1, and the AUC 0-t The ratio was 116:20:1, and the systemic exposure was linearly related to the administered dose. There was no significant difference in the half-life, retention time and clearance rate of the high, middle and low dose groups, as shown in Figure 2.
由于DR303X6在24小时以后血药浓度已经低于检测下限,图2中未有显示。Since the blood concentration of DR303X6 has been lower than the lower limit of detection after 24 hours, it is not shown in Figure 2.
根据上述,DR303X8的体内半衰期长,体内稳定性非常理想。According to the above, DR303X8 has a long half-life in vivo and ideal stability in vivo.
实施例6、抗CLD18A2/HSA/CD3三特异性融合蛋白在PBMC免疫系统人源化BxPC3/hCLDN18.2胰腺癌皮下移植瘤模型中的的抑瘤药效Example 6. Anti-tumor efficacy of anti-CLD18A2/HSA/CD3 trispecific fusion protein in PBMC immune system humanized BxPC3/hCLDN18.2 pancreatic cancer subcutaneous xenograft tumor model
建立PBMC免疫系统人源化BxPC3/hCLDN18.2胰腺癌皮下移植瘤模型,选取雄性NCG小鼠,约7周龄。将BxPC3/hCLDN18.2肿瘤细胞接种在小鼠右侧胁肋部皮下,5x106/小鼠,PBMC来源于正常人外周血,尾静脉注射至荷瘤鼠体内,2x106/小鼠。当平均肿瘤体积达到60mm3左右时分组给药,共分为5组,每组6只动物,分别腹腔注射空白制剂(vehicle,每周2次,连续给药5周)、0.03mg/kg DR303X8(每周2次,连续给药5周)、0.03mg/kg DR303X8(每天1次,连续给药17天)、0.1mg/kg DR303X8(每周2次,连续给药5周)、0.1mg/kg DR303X8(每天1次,连续给药17 天),停药观察。肿瘤体积一周两次测定:采用游标卡尺测定肿瘤的最大长轴(L)和最大宽轴(W),肿瘤体积按如下公式计算:V=L×W2/2,待空白组肿瘤生长至2000mm3或动物状态不足以支持试验时结束试验。To establish a PBMC immune system humanized BxPC3/hCLDN18.2 pancreatic cancer subcutaneous xenograft model, select male NCG mice, about 7 weeks old. BxPC3/hCLDN18.2 tumor cells were inoculated subcutaneously in the right flank of mice, 5x10 6 /mouse, PBMCs were derived from normal human peripheral blood, and injected into tumor-bearing mice by tail vein, 2x10 6 /mouse. When the average tumor volume reached about 60mm3 , the drugs were divided into groups, divided into 5 groups, 6 animals in each group, and blank preparation (vehicle, 2 times a week, continuous administration for 5 weeks), 0.03mg/kg DR303X8 (twice a week, continuous administration for 5 weeks), 0.03mg/kg DR303X8 (once a day, continuous administration for 17 days), 0.1mg/kg DR303X8 (twice a week, continuous administration for 5 weeks), 0.1mg /kg DR303X8 (once a day, continuous administration of 17 Days), drug withdrawal and observation. The tumor volume was measured twice a week: the maximum long axis (L) and maximum width axis (W) of the tumor were measured with a vernier caliper, and the tumor volume was calculated according to the following formula: V=L×W 2 /2, when the tumor in the blank group grew to 2000 mm 3 Or the animal state is not enough to support the test to end the test.
实验结果如图3所示,低浓度给药组和高浓度给药组的DR303X8对PBMC免疫系统人源化BxPC3/hCLDN18.2胰腺癌皮下移植瘤模型均有显著的抑瘤作用。The experimental results are shown in Figure 3, DR303X8 in the low-concentration administration group and high-concentration administration group had significant tumor inhibitory effects on the PBMC immune system humanized BxPC3/hCLDN18.2 pancreatic cancer subcutaneous xenograft tumor model.
实施例7、抗CLD18A2/HSA/CD3三特异性融合蛋白在PBMC免疫系统人源化NUGC4/hCLDN18.2胃癌皮下移植瘤模型中的的抑瘤药效Example 7. Anti-tumor efficacy of anti-CLD18A2/HSA/CD3 trispecific fusion protein in PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft tumor model
建立PBMC免疫系统人源化NUGC4/hCLDN18.2胃癌皮下移植瘤模型,选取雄性NCG小鼠,约7周龄。将NUGC4/hCLDN18.2肿瘤细胞接种在小鼠右侧胁肋部皮下,5x106/小鼠,PBMC来源于正常人外周血,尾静脉注射至荷瘤鼠体内,2x106/小鼠。当平均肿瘤体积达到60mm3左右时分组给药,共分为5组,每组6只动物,分别腹腔注射空白制剂(vehicle,每周2次,连续给药5周)、0.03mg/kg DR303X8(每周2次,连续给药5周)、0.03mg/kg DR303X8(每天1次,连续给药17天)、0.1mg/kg DR303X8(每周2次,连续给药5周)、0.1mg/kg DR303X8(每天1次,连续给药17天)。肿瘤体积一周两次测定:采用游标卡尺测定肿瘤的最大长轴(L)和最大宽轴(W),肿瘤体积按如下公式计算:V=L×W2/2,对照组肿瘤体积均值到达2000mm3时或动物状态不足以支持试验时结束试验。The PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft model was established, and male NCG mice, about 7 weeks old, were selected. NUGC4/hCLDN18.2 tumor cells were inoculated subcutaneously in the right flank of mice, 5x10 6 /mouse, PBMCs were derived from normal human peripheral blood, and injected into tumor-bearing mice by tail vein, 2x10 6 /mouse. When the average tumor volume reached about 60mm3 , the drugs were divided into groups, divided into 5 groups, 6 animals in each group, and blank preparation (vehicle, 2 times a week, continuous administration for 5 weeks), 0.03mg/kg DR303X8 (twice a week, continuous administration for 5 weeks), 0.03mg/kg DR303X8 (once a day, continuous administration for 17 days), 0.1mg/kg DR303X8 (twice a week, continuous administration for 5 weeks), 0.1mg /kg DR303X8 (once a day, continuous administration for 17 days). The tumor volume was measured twice a week: the maximum long axis (L) and maximum width axis (W) of the tumor were measured with a vernier caliper, and the tumor volume was calculated according to the following formula: V=L×W 2 /2, and the average tumor volume of the control group reached 2000mm 3 The test was terminated when the time or the state of the animal was not enough to support the test.
实验结果如图4所示,低浓度给药组和高浓度给药组的DR303X8对PBMC免疫系统人源化NUGC4/hCLDN18.2胃癌皮下移植瘤模型均有显著的抑瘤作用。The experimental results are shown in Figure 4, DR303X8 in the low-concentration administration group and high-concentration administration group had significant tumor inhibitory effects on the PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft tumor model.
实施例8、融合蛋白DR303X8和X910在PBMC免疫系统人源化NUGC4/hCLDN18.2胃癌皮下移植瘤模型中的的抑瘤药效Example 8. Antitumor efficacy of fusion proteins DR303X8 and X910 in PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft tumor model
选用PBMC免疫系统人源化NUGC4/hCLDN18.2胃癌皮下移植瘤模型,选取雄性NCG小鼠,约7周龄。将NUGC4/hCLDN18.2肿瘤细胞接种在小鼠右侧胁肋部皮下,5x106/小鼠;PBMC来源于正常人外周血,尾静脉注射至荷瘤鼠体内,2x106/小鼠。当平均肿瘤体积达到60mm3左右时分组给药,共分为5组,每组6只动物,分别腹腔注射空白制剂(vehicle,每天1次,连续给药29天)、0.01mg/kg DR303X8(每天1次,连续给药29天),0.03mg/kg DR303X8(每周2次,连续给药5次)、0.03mg/kg DR303X8(每天1次,连续给药17天),0.06mg/kg X910(每天1次,连续给药32天)。肿瘤体积一周两次测定:采用游标卡尺测定肿瘤的最大长轴(L)和最大宽轴(W),肿瘤体积按如下公式计算:V=L×W2/2,对照组肿瘤体积均值到达2000mm3时或动物状态不足以支持试验时结束试验。 The PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft model was selected, and male NCG mice were selected, about 7 weeks old. NUGC4/hCLDN18.2 tumor cells were inoculated subcutaneously in the right flank of mice, 5x10 6 /mouse; PBMCs were derived from normal human peripheral blood, and injected into tumor-bearing mice by tail vein, 2x10 6 /mouse. When the average tumor volume reached about 60mm3 , the drugs were divided into groups, divided into 5 groups, with 6 animals in each group, and the blank preparation (vehicle, once a day, continuously administered for 29 days), 0.01 mg/kg DR303X8 ( Once a day, continuous administration for 29 days), 0.03mg/kg DR303X8 (twice a week, continuous administration for 5 times), 0.03mg/kg DR303X8 (once a day, continuous administration for 17 days), 0.06mg/kg X910 (once a day, continuous administration for 32 days). The tumor volume was measured twice a week: the maximum long axis (L) and maximum width axis (W) of the tumor were measured with a vernier caliper, and the tumor volume was calculated according to the following formula: V=L×W 2 /2, and the average tumor volume of the control group reached 2000mm 3 The test was terminated when the time or the state of the animal was not enough to support the test.
实验结果如图5所示,DR303X8对PBMC免疫系统人源化NUGC4/hCLDN18.2胃癌皮下移植瘤模型有明显抑瘤作用。The experimental results are shown in Figure 5, DR303X8 has a significant tumor inhibitory effect on the PBMC immune system humanized NUGC4/hCLDN18.2 gastric cancer subcutaneous xenograft tumor model.
以上所述实施例仅表达了本发明的几种实施方式,其描述较为具体和详细,但并不能因此而理解为对本发明专利范围的限制。应当指出的是,对于本领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干变形和改进,这些都属于本发明的保护范围。因此,本发明专利的保护范围应以所附权利要求为准。同时,在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。 The above-mentioned embodiments only express several implementation modes of the present invention, and the description thereof is relatively specific and detailed, but should not be construed as limiting the patent scope of the present invention. It should be noted that, for those skilled in the art, several modifications and improvements can be made without departing from the concept of the present invention, and these all belong to the protection scope of the present invention. Therefore, the protection scope of the patent for the present invention should be based on the appended claims. Also, all documents mentioned in this application are incorporated by reference in this application as if each were individually incorporated by reference.

Claims (13)

  1. 一种融合蛋白,包括:抗CLD18A2结构域、抗人血清白蛋白结构域和抗CD3结构域;A fusion protein comprising: an anti-CLD18A2 domain, an anti-human serum albumin domain and an anti-CD3 domain;
    所述抗CLD18A2结构域为单域抗体,具有SEQ ID NO:1中第26-33位所示的CDR1,SEQ ID NO:1中第51-57位所示的CDR2,SEQ ID NO:1中第96-109位所示的CDR3;The anti-CLD18A2 structural domain is a single domain antibody with CDR1 shown in positions 26-33 of SEQ ID NO:1, CDR2 shown in positions 51-57 of SEQ ID NO:1, and CDR2 shown in positions 51-57 of SEQ ID NO:1. CDR3 indicated at positions 96-109;
    所述抗人血清白蛋白结构域为单域抗体,具有SEQ ID NO:1中第155-162位所示的CDR1,SEQ ID NO:1中第180-187位所示的CDR2,SEQ ID NO:1中第226-233位所示的CDR3。The anti-human serum albumin domain is a single-domain antibody, having CDR1 shown in positions 155-162 in SEQ ID NO: 1, CDR2 shown in positions 180-187 in SEQ ID NO: 1, and SEQ ID NO : CDR3 shown in positions 226-233 in 1.
  2. 如权利要求1所述的融合蛋白,其特征在于,所述抗CLD18A2结构域为单域抗体,其框架区存在氨基酸序列的P→A突变;较佳地,其FR1区存在P→A突变;更佳地,其P→A突变对应SEQ ID NO:1序列的第14位;和/或The fusion protein according to claim 1, wherein the anti-CLD18A2 domain is a single domain antibody, and its framework region has a P→A mutation in its amino acid sequence; preferably, its FR1 region has a P→A mutation; More preferably, its P→A mutation corresponds to the 14th position of SEQ ID NO:1 sequence; and/or
    所述抗人血清白蛋白结构域为单域抗体,其框架区存在氨基酸序列的P→A突变;较佳地,其FR1区存在P→A突变;更佳地,其P→A突变的氨基酸为对应SEQ ID NO:1序列的第143位的氨基酸。The anti-human serum albumin domain is a single-domain antibody, and its framework region has a P→A mutation in its amino acid sequence; preferably, its FR1 region has a P→A mutation; more preferably, its P→A mutated amino acid It is the 143rd amino acid corresponding to the sequence of SEQ ID NO:1.
  3. 如权利要求1所述的融合蛋白,其特征在于,所述抗CD3结构域为单链抗体;较佳地,其具有SEQ ID NO:1中第279-286位所示的HCDR1,SEQ ID NO:1中第304-313位所示的HCDR2,SEQ ID NO:1中第352-367位所示的HCDR3,SEQ ID NO:1中第419-427位所示的LCDR1,SEQ ID NO:1中第445-447位所示的LCDR2,SEQ ID NO:1中第484-492位所示的LCDR3;和/或The fusion protein according to claim 1, wherein the anti-CD3 domain is a single-chain antibody; preferably, it has HCDR1 shown in positions 279-286 in SEQ ID NO:1, SEQ ID NO HCDR2 shown in No. 304-313 positions in :1, HCDR3 shown in No. 352-367 positions in SEQ ID NO: 1, LCDR1 shown in No. 419-427 positions in SEQ ID NO: 1, SEQ ID NO: 1 and/or
    所述的抗CD3结构域位于抗人血清白蛋白结构域是N端或C端。The anti-CD3 structural domain is located at the N-terminal or C-terminal of the anti-human serum albumin structural domain.
  4. 如权利要求1~3任一所述的融合蛋白,其特征在于,所述抗CLD18A2结构域的氨基酸序列如SEQ ID NO:1中第1~120位所示;The fusion protein according to any one of claims 1 to 3, wherein the amino acid sequence of the anti-CLD18A2 domain is shown in positions 1 to 120 in SEQ ID NO:1;
    所述抗人血清白蛋白结构域的氨基酸序列如SEQ ID NO:1中第130~244位所示;和/或The amino acid sequence of the anti-human serum albumin domain is shown in the 130th to 244th positions in SEQ ID NO: 1; and/or
    所述抗CD3结构域的氨基酸序列如SEQ ID NO:1中第254~502位所示。The amino acid sequence of the anti-CD3 domain is shown in the 254th to 502nd positions in SEQ ID NO:1.
  5. 如权利要求1所述的融合蛋白,其特征在于,所述抗CLD18A2结构域、抗人血清白蛋白结构域和抗CD3结构域的氨基酸序列直接连接,或通过连接子连接,所述的连接子相同或不同;较佳地,所述的融合蛋白的氨基酸序列如SEQ ID NO:1所示。 The fusion protein according to claim 1, wherein the amino acid sequences of the anti-CLD18A2 domain, the anti-human serum albumin domain and the anti-CD3 domain are directly connected, or connected by a linker, and the linker The same or different; preferably, the amino acid sequence of the fusion protein is shown in SEQ ID NO:1.
  6. 一种多核苷酸或含有该多核苷酸的表达构建体,其特征在于,所述多核苷酸编码权利要求1~5任一所述的融合蛋白。A polynucleotide or an expression construct containing the polynucleotide, characterized in that the polynucleotide encodes the fusion protein according to any one of claims 1-5.
  7. 一种融合蛋白的表达系统,所述表达系统含有权利要求6所述的表达构建体或其基因组中整合有外源的权利要求6所述的多核苷酸。A fusion protein expression system, the expression system contains the expression construct of claim 6 or the exogenous polynucleotide of claim 6 integrated in its genome.
  8. 制备权利要求1~5任一所述的融合蛋白的方法,其特征在于,所述方法包括:利用权利要求7所述的表达系统表达所述的融合蛋白;较佳地,所述方法还包括:分离出所述的融合蛋白。The method for preparing the fusion protein according to any one of claims 1 to 5, characterized in that the method comprises: expressing the fusion protein using the expression system according to claim 7; preferably, the method further comprises : the fusion protein is isolated.
  9. 一种改造CLD18A2结合分子以提高其疗效的方法,其特征在于,所述方法包括:以抗CLD18A2结构域作为CLD18A2结合分子,将其与抗人血清白蛋白结构域和抗CD3结构域连接;A method for modifying a CLD18A2 binding molecule to improve its curative effect, characterized in that the method comprises: using an anti-CLD18A2 domain as a CLD18A2 binding molecule, linking it to an anti-human serum albumin domain and an anti-CD3 domain;
    所述抗CLD18A2结构域为单域抗体,具有SEQ ID NO:1中第26-33位所示的CDR1,SEQ ID NO:1中第51-57位所示的CDR2,SEQ ID NO:1中第96-109位所示的CDR3;The anti-CLD18A2 structural domain is a single domain antibody with CDR1 shown in positions 26-33 of SEQ ID NO:1, CDR2 shown in positions 51-57 of SEQ ID NO:1, and CDR2 shown in positions 51-57 of SEQ ID NO:1. CDR3 indicated at positions 96-109;
    所述抗人血清白蛋白结构域为单域抗体,具有SEQ ID NO:1中第155-162位所示的CDR1,SEQ ID NO:1中第180-187位所示的CDR2,SEQ ID NO:1中第226-233位所示的CDR3;The anti-human serum albumin domain is a single-domain antibody, having CDR1 shown in positions 155-162 in SEQ ID NO: 1, CDR2 shown in positions 180-187 in SEQ ID NO: 1, and SEQ ID NO : CDR3 shown in positions 226-233 in 1;
    所述的抗CD3结构域位于抗人血清白蛋白结构域是N端或C端。The anti-CD3 structural domain is located at the N-terminal or C-terminal of the anti-human serum albumin structural domain.
  10. 如权利要求9所述的方法,其特征在于,所述抗CLD18A2结构域为单域抗体,对其框架区进行氨基酸序列的P→A突变;较佳地,对其FR1区进行P→A突变;更佳地,其P→A突变对应SEQ ID NO:1序列的第14位;和/或The method according to claim 9, wherein the anti-CLD18A2 domain is a single domain antibody, and the framework region thereof is subjected to a P→A mutation of the amino acid sequence; preferably, a P→A mutation is performed on the FR1 region ; More preferably, its P → A mutation corresponds to the 14th position of SEQ ID NO: 1 sequence; and/or
    所述抗人血清白蛋白结构域为单域抗体,对其框架区进行氨基酸序列的P→A突变;较佳地,对其FR1区进行P→A突变;更佳地,其P→A突变的氨基酸为对应SEQ ID NO:1序列的第143位的氨基酸;和/或The anti-human serum albumin domain is a single-domain antibody, and its framework region is subjected to a P→A mutation in its amino acid sequence; preferably, its FR1 region is subjected to a P→A mutation; more preferably, its P→A mutation The amino acid is the 143rd amino acid corresponding to the sequence of SEQ ID NO:1; and/or
    所述抗CD3结构域为单链抗体;较佳地,其具有SEQ ID NO:1中第279-286位所示的HCDR1,SEQ ID NO:1中第304-313位所示的HCDR2,SEQ ID NO:1中第352-367位所示的HCDR3,SEQ ID NO:1中第419-427位所示的LCDR1,SEQ ID NO:1中第445-447位所示的LCDR2,SEQ ID NO:1中第484-492位所示的LCDR3。The anti-CD3 structural domain is a single-chain antibody; preferably, it has HCDR1 shown in positions 279-286 in SEQ ID NO: 1, HCDR2 shown in positions 304-313 in SEQ ID NO: 1, SEQ ID NO: 1 HCDR3 shown in No. 352-367 in ID NO:1, LCDR1 shown in No. 419-427 in SEQ ID NO:1, LCDR2 shown in No. 445-447 in SEQ ID NO:1, SEQ ID NO : LCDR3 shown in bits 484-492 in 1.
  11. 权利要求1~5任一所述的融合蛋白、权利要求7所述的表达系统或其培 养物在制备缓解或治疗CLD18A2阳性细胞相关疾病的药物中的用途;较佳地,所述CLD18A2阳性细胞相关疾病包括肿瘤;较佳地,所述肿瘤包括:胰腺癌、胃癌、食管癌、肺癌、卵巢癌、乳腺癌、结肠直肠癌、肝癌、胆囊癌或头颈癌。The fusion protein according to any one of claims 1 to 5, the expression system according to claim 7 or its culture The use of nutrients in the preparation of drugs for alleviating or treating CLD18A2 positive cell-related diseases; preferably, the CLD18A2 positive cell-related diseases include tumors; preferably, the tumors include: pancreatic cancer, gastric cancer, esophageal cancer, lung cancer , ovarian, breast, colorectal, liver, gallbladder, or head and neck cancer.
  12. 一种药物组合物,其特征在于,所述药物组合物包括:A kind of pharmaceutical composition, is characterized in that, described pharmaceutical composition comprises:
    权利要求1~5任一所述的融合蛋白;或The fusion protein of any one of claims 1-5; or
    权利要求7所述的融合蛋白的表达系统,或其培养物。The expression system of the fusion protein according to claim 7, or its culture.
  13. 一种药盒,其特征在于,所述药盒包括:容器或包装,以及位于其中的:A kind of medicine box, it is characterized in that, described medicine box comprises: container or packing, and be positioned at wherein:
    权利要求1~5任一所述的融合蛋白;或The fusion protein according to any one of claims 1-5; or
    权利要求7所述的融合蛋白的表达系统,或其培养物;或The expression system of the fusion protein according to claim 7, or its culture; or
    权利要求12所述的药物组合物。 The pharmaceutical composition of claim 12.
PCT/CN2023/072066 2022-01-14 2023-01-13 Three-target anti-tumor drug, and preparation method therefor and use thereof WO2023134742A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210042780.0 2022-01-14
CN202210042780 2022-01-14

Publications (1)

Publication Number Publication Date
WO2023134742A1 true WO2023134742A1 (en) 2023-07-20

Family

ID=87280141

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/072066 WO2023134742A1 (en) 2022-01-14 2023-01-13 Three-target anti-tumor drug, and preparation method therefor and use thereof

Country Status (1)

Country Link
WO (1) WO2023134742A1 (en)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103619878A (en) * 2011-06-23 2014-03-05 埃博灵克斯股份有限公司 Serum albumin binding proteins
CN110913908A (en) * 2017-05-12 2020-03-24 哈普恩治疗公司 MSLN-targeting trispecific proteins and methods of use
CN111234027A (en) * 2015-05-21 2020-06-05 哈普恩治疗公司 Trispecific binding proteins and methods of use
CN111434692A (en) * 2019-01-15 2020-07-21 浙江道尔生物科技有限公司 anti-C L D18A2 nano antibody and application thereof
CN112512581A (en) * 2018-08-03 2021-03-16 安进研发(慕尼黑)股份有限公司 Antibody constructs directed against CLDN18.2 and CD3
WO2021091906A1 (en) * 2019-11-04 2021-05-14 Amgen Inc. Methods for treating leukemia
WO2021181396A1 (en) * 2020-03-12 2021-09-16 Biond Biologics Ltd. Shedding blocking agents with increased stability
CN113461824A (en) * 2020-03-31 2021-10-01 普米斯生物技术(珠海)有限公司 Platform for constructing multispecific antibody
WO2021231434A1 (en) * 2020-05-12 2021-11-18 Harpoon Therapeutics, Inc. Psma targeting tritacs and methods of use
WO2022011531A1 (en) * 2020-07-14 2022-01-20 浙江道尔生物科技有限公司 Anti-cld18a2 single-domain antibody

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103619878A (en) * 2011-06-23 2014-03-05 埃博灵克斯股份有限公司 Serum albumin binding proteins
CN111234027A (en) * 2015-05-21 2020-06-05 哈普恩治疗公司 Trispecific binding proteins and methods of use
CN110913908A (en) * 2017-05-12 2020-03-24 哈普恩治疗公司 MSLN-targeting trispecific proteins and methods of use
CN112512581A (en) * 2018-08-03 2021-03-16 安进研发(慕尼黑)股份有限公司 Antibody constructs directed against CLDN18.2 and CD3
CN111434692A (en) * 2019-01-15 2020-07-21 浙江道尔生物科技有限公司 anti-C L D18A2 nano antibody and application thereof
WO2021091906A1 (en) * 2019-11-04 2021-05-14 Amgen Inc. Methods for treating leukemia
WO2021181396A1 (en) * 2020-03-12 2021-09-16 Biond Biologics Ltd. Shedding blocking agents with increased stability
CN113461824A (en) * 2020-03-31 2021-10-01 普米斯生物技术(珠海)有限公司 Platform for constructing multispecific antibody
WO2021231434A1 (en) * 2020-05-12 2021-11-18 Harpoon Therapeutics, Inc. Psma targeting tritacs and methods of use
WO2022011531A1 (en) * 2020-07-14 2022-01-20 浙江道尔生物科技有限公司 Anti-cld18a2 single-domain antibody

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ZHANG, JIANWEI ET AL.: "Evaluation and Reflection on Claudin 18.2 Targeting Therapy in Advanced Gastric Cancer.", CHINESE JOURNAL OF CANCER RESEARCH., vol. 32, no. 2, 2020-12-31, XP055929148, DOI: 10.21147/j.issn.1000-9604.2020.02.13 *
ZHU, GUOYUN ET AL.: "Targeting CLDN18.2 by CD3 Bispecific and ADC Modalities for the Treatments of Gastric and Pancreatic Cancer.", SCIENTIFIC REPORTS., vol. 9, 2019-06-10, XP055630964, DOI: 10.1038/s41598-019-44874-0 *

Similar Documents

Publication Publication Date Title
CN111683970B (en) C-KIT binding agents
CA3092456C (en) Anti-tigit antibody and use thereof
JP6432121B2 (en) PDL-1 antibody, pharmaceutical composition thereof and use thereof
CN105121474B (en) Fusion immunomodulatory protein and preparation method thereof
JP7011574B2 (en) Antibody constructs against FLT3 and CD3
EP2986630B1 (en) Novel bispecific binding molecules with antitumoral activity
KR101721301B1 (en) Bispecific antigen binding molecules
AU2003205716B2 (en) FGFR agonists
EP3760642A1 (en) Human antigen binding proteins that bind beta-klotho, fgf receptors and complexes thereof
SG177193A1 (en) Human antibodies to human delta like ligand 4
WO2018166507A1 (en) Novel recombinant bifunctional fusion protein, preparation method therefor and use thereof
EP2920210B1 (en) Recombinant bispecific antibody binding to cd20 and cd95
CN110835374A (en) anti-CCR 8 × CTLA-4 bispecific antibody and application thereof
TW202126700A (en) A fusion protein and uses thereof
WO2021078123A1 (en) RECOMBINANT PROTEIN TARGETING PD-1 AND TGFβ
CN114380915A (en) anti-CD 73 antibodies and uses thereof
JP2022553908A (en) PD1 and VEGFR2 double binding agents
WO2023134742A1 (en) Three-target anti-tumor drug, and preparation method therefor and use thereof
WO2022174781A1 (en) Multi-domain fusion protein and use thereof
CN111788229A (en) CSF1R binding agents
US11629186B2 (en) Anti-CCL8 antibodies and uses thereof
EP3082858B1 (en) Adrenomedullin binder for use in therapy of cancer
CN115304680A (en) Preparation and application of bispecific cell adaptor molecule constructed based on Pep42
WO2017211278A1 (en) Antibody fusion proteins for drug delivery
WO2022174452A1 (en) Bifunctional fusion protein having anticancer activity

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: 23740083

Country of ref document: EP

Kind code of ref document: A1