WO2021203608A1 - Vegfr2-targeted metastatic cancer vaccine - Google Patents

Vegfr2-targeted metastatic cancer vaccine Download PDF

Info

Publication number
WO2021203608A1
WO2021203608A1 PCT/CN2020/109970 CN2020109970W WO2021203608A1 WO 2021203608 A1 WO2021203608 A1 WO 2021203608A1 CN 2020109970 W CN2020109970 W CN 2020109970W WO 2021203608 A1 WO2021203608 A1 WO 2021203608A1
Authority
WO
WIPO (PCT)
Prior art keywords
vegfr2
cells
fusion protein
protein
cell
Prior art date
Application number
PCT/CN2020/109970
Other languages
French (fr)
Chinese (zh)
Inventor
齐海龙
王晓芳
严小娥
李日勇
罗天明
谢皇帆
刘德芳
孙忠杰
Original Assignee
诺未科技(北京)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 诺未科技(北京)有限公司 filed Critical 诺未科技(北京)有限公司
Publication of WO2021203608A1 publication Critical patent/WO2021203608A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00113Growth factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to the field of cancer immunotherapy and prevention, in particular to a metastatic cancer vaccine targeting VEGFR2.
  • Tumor metastasis is the main cause of cancer death. More than 90% of cancer death rates are caused by metastasis, and the formation of tumor blood vessels is essential for tumor growth and metastasis (Valastyan and Weinberg). Tumor blood vessels provide sufficient oxygen and nutrients for tumor growth and metastasis. The formation of new blood vessels is a necessary step for tumor blood vessel formation.
  • the neovascularization theory believes that tumors induce the formation of new blood vessels from existing blood vessels. Based on this theory, researchers have developed a series of drugs that target tumor neovascularization.
  • the current FDA-approved anti-angiogenesis drugs are mainly divided into three categories: monoclonal antibodies, receptor tyrosine kinase inhibitors, and fusion peptides. .
  • bevacizumab, sorafenib, and the fusion protein of the extracellular domain of the VEGFR1/2 protein used in the treatment of metastatic cancer can effectively improve the patient's progression-free survival (Cascone and Heymach).
  • progression-free survival Cascone and Heymach.
  • anti-angiogenic drugs Taking bevacizumab as an example, frequent parenteral administration, serious side effects, obvious drug resistance, and tumor rebound progression after metastasis are needed. These problems need to be resolved urgently.
  • vascular endothelial growth factor is the most important regulator of neovascularization, and its function is mainly through the binding of three receptor tyrosine kinases VEGFR1/2/3.
  • VEGFR1 has high affinity for VEGF but low intracellular enzyme activity, which has a negative feedback inhibitory effect on the formation of new blood vessels.
  • VEGFR3 is mainly expressed in adult lymphatic vessels, while VEGFR2 is the main mediator of VEGF-induced new blood vessels. Promoting factors (Dong et al.) that produce and increase endothelial cell permeability.
  • VEGFR2 knockout mice developed to 8.5-9.5 days and died due to impaired angiogenesis (Shalaby et al.).
  • VEGFR2 is highly expressed in tumor-associated endothelial cells in tumor tissues, and its expression is much higher than that of normal vascular cells. In addition to tumor-associated endothelial cells, VEGFR2 is also highly expressed in certain types of tumor cells, such as B cells Lymphoma, breast cancer, lung cancer, bladder cancer and melanoma, etc. (Decaussin et al., 1999; El-Obeid et al., 2004; Gakiopoulou-Givalou et al., 2003; Kranz et al., 1999), thus VEGFR2 is an ideal target for tumor therapy.
  • the bevacizumab and sorafenib are anti-tumor drugs developed with VEGFR2 as the target.
  • VEGFR2 vascular endothelial growth factor receptor 2
  • VEGFR2 is an embryonic tumor-associated antigen, it can be recognized and highly expressed during the body’s cellular immune development.
  • Affinity-bound T cells have been eliminated by negative selection in the thymus (Houghton et al., 2007). Only T cells with low affinity for VEGFR2 survive the selection of the thymus, but these T cells cannot be activated by the body's own VEGFR2. Therefore, although tumor cells highly express VEGFR2, the body itself cannot produce effector T cells that can effectively recognize tumor cells with high expression of VEGFR2, thereby inhibiting angiogenesis and tumor growth. This phenomenon is called immune tolerance.
  • VEGFR2169-177: RFVPDGNRI and VEGFR2773-781: VLAMFFWLL, respectively) are used to immunize patients with advanced metastatic colorectal cancer and nerve fibers
  • Tumor patients conducted phase I/II clinical trials, the results showed that after stimulation with peptide vaccines, the increased proportion of VEGFR2-specific cytotoxic T cells (CTLs) can inhibit tumor angiogenesis, increase the number of CD8+ T cells in the tumor, and eliminate VEGFR2-positive tumor cells reduce tumor burden and improve the overall survival rate of patients (Hazama et al., 2014) and (Tamura et al., 2019).
  • CTLs cytotoxic T cells
  • HLA-A2 and A24 Due to the HLA-restricted T cell immune response and polypeptide immunity generally cannot effectively cause antibody-mediated humoral immune responses, HLA- A2 and HLA-A24 restricted VEGFR immunogenic peptides may not have any effect on other HLA subtypes. Moreover, the immunological peptides used without epitope optimization can cause a limited degree of immune response, which may limit the effectiveness of this type of vaccine. In summary, it is necessary to design a preventive and therapeutic vaccine that targets VEGFR2 that can effectively break immune tolerance, antagonize angiogenesis and inhibit the formation of metastatic cancer foci through cell-specific immune responses.
  • Antigen presenting cells refer to a type of cells that can process antigens and present antigen peptides to T cells in the form of antigen peptide-MHC molecular complexes, and play an important role in the body's immune recognition, immune response and immune regulation.
  • Dendritic cell (DC) cell is a type of antigen-presenting cell in the body, and it is the only antigen-presenting cell in the body that can present antigen peptides to naive T cells and induce T cell activation and proliferation. , Is the initiator of the body's adaptive immune response. According to morphology and function, DC cells can be divided into two categories: classic DC and plasma cell-like DC. Plasma cell-like DCs mainly mediate the adaptive immune response after virus infection.
  • Classical DCs have the strongest ability to process and present antigens, which mediate the elimination of dead cells in the body and the recognition of tumor cells (Carreno et al., 2017; Minetto et al., 2019). Therefore, more effective presentation of tumor antigens to classic DCs is the key to the success of the vaccine. Studies have found that classic DCs express a class of chemokine XCL1 receptors, so XCL1 molecules can efficiently bind to classic DC cells (Dorner et al., 2009) .
  • the present invention provides a metastatic cancer vaccine targeting VEGFR2.
  • the invention improves the efficiency of phagocytosis, processing and presentation of the VEGFR2 protein by DC cells by fusion expression of VEGFR2 protein which promotes tumor angiogenesis and DC cell ligand XCL1, and improves the effect of inhibiting tumor growth.
  • the protein expressed by the nucleic acid vaccine of the present invention can effectively bind to DC cells, induce VEGFR2-specific T cell responses, and significantly inhibit the growth of melanomas that highly express VEGFR2 in a variety of models.
  • the fusion protein provided by the present invention includes the extracellular region of VEFGR2 that promotes tumor angiogenesis that can induce a specific CD8+T response or the extracellular region of the optimized form of binding epitope of MHC class I molecules; linkers; and specific binding with antigen crossover Human or mouse XCL1 protein of DC cells that present the ability;
  • VEFGR2 The extracellular region of VEFGR2 that promotes tumor angiogenesis that can induce a specific CD8+T response or its MHC class I molecule binding epitope optimized form has the extracellular region as shown in SEQ ID No. 1 or 2.
  • the fusion protein includes the sequence shown in amino acids 1 to 889 in SEQ ID Nos. 7 and 8.
  • the present invention also provides a nucleotide encoding the fusion protein, which has
  • (III) A nucleotide sequence that encodes the same protein as the nucleotide sequence of (I) or (II), but is different from the nucleotide sequence of (I) or (II) due to the degeneracy of the genetic code;
  • the nucleotide sequence shown in (II) or (III) has the same or similar function;
  • the present invention also provides a recombinant expression vector, including a vector and the fusion protein or the nucleotide of the present invention.
  • the vector includes a mammalian cell expression vector or an insect rod-shaped cell expression vector; the vector includes pcDNA3.1(+), pcDNA3.1(-), pFastbac1-dual-MBP .
  • the present invention also provides a recombinant strain or cell comprising the fusion protein or the nucleotide of the present invention.
  • the present invention also provides a vaccine for preparing metastatic cancer or melanoma with high expression of VEGFR2 in the fusion protein, the nucleotide, the recombinant expression vector or the recombinant strain or cell. Or application in the preparation of medicines for preventing and/or treating metastatic cancer or melanoma with high expression of VEGFR2.
  • the metastatic cancer includes liver cancer, colorectal cancer or lung cancer.
  • the present invention also provides a vaccine for metastatic cancer or melanoma with high expression of VEGFR2, including the fusion protein of the present invention, the nucleotide, the recombinant expression vector and the pharmaceutical Acceptable carriers, excipients and/or adjuvants.
  • the present invention also provides drugs for the prevention and/or treatment of metastatic cancer or melanoma with high expression of VEGFR2, including the fusion protein of the present invention, the nucleotide, and the recombinant Expression vector and pharmaceutically acceptable excipients.
  • the vaccine provided by the present invention is composed of the extracellular region of the vascular endothelial cell growth factor receptor VEGFR2 that can cause a specific CD8+ T cell immune response and the extracellular region of the optimized form of MHC class I molecule binding epitope, and is expressed by fusion at its N-terminus
  • the protein composed of the XCL1 molecule of DC that specifically binds to the antigen cross-presenting ability contains the nucleic acid of the fusion protein and the carrier where the nucleic acid is located, and contains the fusion protein of the present invention.
  • the nucleic acid and the carrier are used in the prevention and treatment of melanoma and metastatic cancer. Applications.
  • the invention improves the efficiency of phagocytosis, processing and presentation of the VEGFR2 protein by DC cells by fusion expression of VEGFR2 protein which promotes tumor angiogenesis and DC cell ligand XCL1, and improves the effect of inhibiting tumor growth.
  • the protein expressed by the nucleic acid vaccine of the present invention can effectively bind to DC cells, induce VEGFR2-specific T cell responses, and significantly inhibit the growth of melanomas that highly express VEGFR2 in a variety of models.
  • Figure 1 shows the vector map of the nucleotide encoding the fusion protein
  • Figure 1A shows the CMV promoter followed by the XCL1 sequence followed by the VEGFR2 wild-type extracellular region for the nucleic acid vaccine coding sequence
  • Figure 1B shows the CMV promoter followed by the XCL1 sequence Connecting VEGFR2MHCI molecules to optimize the mutant extracellular region nucleic acid vaccine coding sequence
  • Figure 1C shows the CMV promoter followed by the XCL1 secretion signal peptide sequence followed by the VEGFR2 extracellular region for nucleic acid vaccine coding sequence
  • Figure 1D shows pFASTbac-dual-MBP -XCL1-VEGFR2 wild-type fusion protein vaccine purification vector
  • Figure 1E shows the pFASTbac-dual-MBP-XCL1-VEGFR2 mutant fusion protein vaccine purification vector
  • Figure 1F shows the pFASTbac-dual-MBP-VEGFR2 wild-type protein vaccine purification vector
  • Figure 2 shows the results of the three-dimensional structure prediction of the fusion protein; input the nucleotide sequence encoding the fusion protein into the website http://raptorx.uchicago.edu/ to predict the three-dimensional structure of the fusion protein, showing the XCL1-VEGFR2 fusion protein space Structure diagram
  • Figure 3 shows the detection of the expression of the nucleic acid encoding the fusion protein in HEK293T cells; after 48 hours of transfection of the plasmid vector carrying the nucleic acid encoding the fusion protein into HEK292T cells, Western blotting was used to detect the C-terminal with a Flag tag The expression of nucleotides encoding the fusion protein;
  • Figure 4 shows the detection of the fusion protein purified from insect rod-shaped cells SF9; Coomassie brilliant blue staining is used to detect the purification of XCL1-VEGFR2 secreted and expressed from insect rod-shaped cells;
  • Figure 5 shows that the fusion protein can effectively bind to MHCII+CD11c+CD8a+ antigen cross-presenting DC cells; the Flag-tagged SF9 cell-purified protein is incubated with the spleen cells enriched with CD11C magnetic beads, and Flag fluorescent antibody is used to detect the fusion protein and MHCII+CD11c+CD8a+ antigen cross-presenting DC cell binding situation; among them, Figure 5 (A) shows the fusion protein binding DC cell flow analysis diagram; Figure 5 (B) shows the statistics of the proportion of fusion protein binding DC cells in 5 (A) diagram;
  • Figure 6 shows the detection of the chemotactic effect of the fusion protein on MHCII+CD11c+CD8a+ antigen cross-presenting DC cells; using the Transwell test, spleen cells containing MHCII+CD11c+CD8a+ antigen cross-presenting DC cells are placed on the upper layer, and SF9 insect cells will be removed Place the purified fusion protein in the lower layer and observe the number of upper MHCII+CD11c+CD8a+ antigen cross-presenting DC cells entering the lower layer after 2 hours; the results show that the fusion protein has a chemotactic effect on MHCII+CD11c+CD8a+ antigen cross-presenting DC cells; , Figure 6(A) shows a flow cytometric analysis chart of fusion protein chemotactic DC cells; Figure 6(B) shows a statistical analysis chart of the proportion of 6(A) fusion protein chemotactic DC cells;
  • Figure 7 shows the detection of VEGFR2 (KDR) expression of melanoma cell line B16 cells after tumorigenesis; tumor cells are inoculated subcutaneously, after tumorigenesis, the tumor tissue and surrounding skin tissue are taken to extract total RNA, and real-time fluorescent quantitative PCR is used to expand
  • the augmentation technique detects the expression of VEGFR2 in the tumor and surrounding skin, and the results show that the expression of VEGFR2 in the tumor tissue is significantly higher than that in the surrounding normal skin;
  • Figure 8 shows the growth of mouse melanoma after immunization with the fusion gene encoding XCL1-VEGFR2/XCL1-VEGFR2 mutant fusion protein;
  • Figure 8A shows the fusion gene immunization method and the immunization process diagram;
  • Figure 8B shows the melanoma after inoculation , A graph of tumor growth curve made by measuring the volume of the tumor;
  • Figure 8C shows that the mice were sacrificed when the tumor in the control group grew to the ethical end point of 2000 mm 3 and the tumor size comparison diagram was dissected;
  • Figure 9 shows an analysis diagram of the proportion of granzyme B-positive CD8a T cells in lymphocytes infiltrating the mouse melanoma after immunization with a fusion gene encoding XCL1-VEGFR2 fusion protein;
  • Figure 10 shows the use of the fusion protein to immunize mice, and then the spleen cells are taken into the mice that were intravenously injected with melanoma cells to detect the formation of simulated melanoma metastatic nodules in the lungs;
  • Figure 10A shows the fusion protein immunized by intravenous injection Method effect diagram;
  • Figure 10B shows a diagram of mouse lung nodules after adoptive treatment with fusion protein immunization.
  • the present invention discloses a metastatic cancer vaccine targeting VEGFR2.
  • Those skilled in the art can learn from the content of this article and appropriately improve the process parameters to achieve it.
  • all similar replacements and modifications are obvious to those skilled in the art, and they are all deemed to be included in the present invention.
  • the method and application of the present invention have been described through the preferred embodiments. It is obvious that relevant persons can make changes or appropriate changes and combinations to the methods and applications described herein without departing from the content, spirit and scope of the present invention to achieve and Apply the technology of the present invention.
  • the present invention uses the amino acid sequence of the extracellular region of VEGFR2 as an immunogen to facilitate the purification of soluble protein vaccines.
  • the vaccine of the present invention can effectively induce specific T cell immune responses against VEGFR2 in humans or mice, breaking The immune tolerance of the original tumor-associated antigen VEGFR2 can effectively directly eliminate tumor cells expressing VEGFR2, and indirectly inhibit tumor growth and the formation of metastases by inhibiting tumor angiogenesis.
  • the vaccine consists of 1) the extracellular region of vascular endothelial growth factor receptor 2 (VEFGR2) that promotes tumor angiogenesis and can induce a specific CD8+T response And its MHC class I molecule binding epitope optimized form extracellular region, and 2) N-terminal fusion expresses the human or mouse XCL1 protein composition that specifically binds to DC cells with antigen cross-presentation ability.
  • VEGFR2 vascular endothelial growth factor receptor 2
  • SEQ ID No. 1 and SEQ ID No. 2 are composed of amino acids 20-764, and the N-terminus is expressed through a linker fusion that specifically binds human or human DC cells with antigen cross-presentation ability.
  • Mouse XCL1 is composed of the sequence shown in amino acids 1-114 of SEQ ID No. 3 or SEQ ID No. 4.
  • the vaccine for metastatic cancer includes any sequence of amino acids 1-889 in SEQ ID No. 7 and SEQ ID No. 8.
  • the nucleic acid vaccine consists of 1) encoding the extracellular vascular endothelial growth factor receptor 2 (VEFGR2) that promotes tumor angiogenesis and can induce a specific CD8+T response. Region and the nucleotide sequence of the extracellular region of the optimized form of binding epitope of MHC class I molecules, and 2) the N-terminal fusion expressing the nucleoside of human or mouse XCL1 protein that specifically binds to DC cells with antigen cross-presenting ability through linker fusion Acid sequence composition. That is, it encodes the 20-764 amino acid sequence of SEQ ID No. 1 and SEQ ID No. 2 and the human or mouse XCL1 sequence that specifically binds to DC cells with antigen cross-presentation ability, that is, SEQ ID No. 3 or SEQ ID No. 4 in the amino acid sequence of positions 1-114.
  • VEFGR2 extracellular vascular endothelial growth factor receptor 2
  • the sequence encoding VEFGR2 that promotes tumor angiogenesis and human or murine XCL1 that specifically binds to DC cells with antigen cross-presenting ability includes the nucleotides in SEQ ID No. 11-SEQ ID No. 16 sequence.
  • the present invention also provides a recombinant expression vector carrying the nucleic acid vaccine, which contains the fusion protein vaccine of the present invention and the nucleotide sequence and vector corresponding to the vaccine vaccine.
  • the vector can be a mammalian cell expression vector or an insect rod-shaped cell expression vector.
  • the vector can be pcDNA3.1(+), pcDNA3.1(-), pFastbac1-dual-MBP.
  • the present invention finally provides a vaccine for metastatic cancer, including the fusion protein vaccine, nucleic acid vaccine and recombinant expression vector.
  • the vaccine of the present invention provides a method for preventing recurrence or treatment for postoperative metastatic cancers such as liver cancer, colorectal cancer, lung cancer, and melanoma with high expression of VEGFR2. Including the administration of the vaccine of the present invention for other indications.
  • the present invention also provides the application of the vaccine, nucleic acid vaccine and vector in preparing liver cancer, colorectal cancer, lung cancer and other metastatic cancers after surgery and melanoma vaccines with high expression of VEGFR2.
  • the present invention also provides the vaccine, nucleic acid vaccine and carrier for treating melanoma.
  • the vaccine or drug of the present invention includes the fusion protein, the nucleic acid, the recombinant expression vector, and optionally pharmaceutically acceptable carriers, excipients and/or adjuvants.
  • the present invention provides a vaccine for metastatic cancer
  • the vaccine is optimized by the extracellular region of the vascular endothelial cell growth factor receptor VEGFR2 that can cause a specific CD8+ T cell immune response and MHC class I molecule binding epitopes.
  • the extracellular region is fused at its N-terminus to express a protein composed of XCL1 molecules that specifically bind to the antigen cross-presenting ability, the nucleic acid containing the fusion protein and the vector in which the nucleic acid is located, including the fusion protein of the present invention, the nucleic acid and the vector Application in the prevention and treatment of melanoma and metastatic cancer.
  • the invention improves the efficiency of phagocytosis, processing and presentation of the VEGFR2 protein by DC cells by fusion expression of VEGFR2 protein which promotes tumor angiogenesis and DC cell ligand XCL1, and improves the effect of inhibiting tumor growth.
  • the protein expressed by the nucleic acid vaccine of the present invention can effectively bind to DC cells, induce VEGFR2-specific T cell responses, and significantly inhibit the growth of melanomas that highly express VEGFR2 in a variety of models.
  • the raw materials and reagents involved in the present invention can be purchased from the market.
  • Example 1 The design scheme of fusion protein vaccine antigen and the construction and preparation of mammalian cell and insect rod-shaped cell expression plasmids
  • the fusion protein XCL1-VEGFR2 was constructed according to the amino acid sequence of mouse VEGFR2 (SEQ ID No.1 and SEQ ID No.2) and mouse XCL1 (SEQ ID No.4), in order to promote the translation of nucleic acid expressing the fusion protein in vivo
  • the fusion protein can be effectively secreted outside the cell and chemotactic MHC-II+CD11c+CD8a+ antigen cross-presenting DC cells.
  • the signal peptide for membrane positioning is removed (SEQ ID No. 6).
  • the final amino acid sequence of the fusion protein (SEQ ID No. 7 and SEQ ID No. 8).
  • the first component of this expression system is the pFastBac strain used to clone the target gene. Based on the selection of the pFastBac strain, the expression gene is controlled by the PH or p10 promoter, and the high-level expression expression frame in insect cells is surrounded by the left and right arms of Tn7, which contains a gentamicin resistance point, forming a miniature Tn7.
  • the second component of this expression system is the DH10Bac strain of E. coli. DH10Bac cells contain viral plasmids with mini-attTn7 target sites and helper plasmids.
  • the virus strain (bacmid) is amplified in the kana-resistant plasmid DH10Bac, and when the coloring substance X-gal and the inducer IPTG are present, it can supplement the lack of chromosome LacZ to form blue spots.
  • micro-Tn7 into the micro-attTn7 attachment site will interfere with the expression of LacZa peptide, so the clone containing the recombinant plasmid is white on the blue background, so choose the white spot analysis.
  • the true leukoplakia clone is relatively large, so in order to avoid false positive selection, choose the largest and most independent leukoplakia.
  • the experiment finally obtained the recombinant plasmid that cloned the target gene into pFastBac HTB; the recombinant plasmid pFastBac HTB transformed into DH10Bac competent cells: Pick the target plaque (white spot): 48 hours later, until the blue and white spots can be clearly distinguished, choose the largest The most independent leukoplakia 2 to 3, use 5ml LB medium (50 ⁇ g/ml kanamycin, 10 ⁇ g/ml tetracycline, 7 ⁇ g/ml gentamicin) shake at 37°C, 220rpm for 8-12 hours, Until the bacterial liquid is turbid, then use this bacterial liquid to extract Bacmid.
  • 5ml LB medium 50 ⁇ g/ml kanamycin, 10 ⁇ g/ml tetracycline, 7 ⁇ g/ml gentamicin
  • XCL1 is a chemokine, and the normal function of its chemotactic function needs to maintain a complete spatial structure.
  • XCL1 still retains its own spatial structure after the fusion of XCL1 and the extracellular domain of VEGFR2
  • a 5-amino acid Flag tag composed of DDDDK to the C-terminus of the fusion protein, so that the Flag tag antibody can be used to detect the expression of the fusion protein.
  • FIG. 3 shows that the empty vector (Vector) has no protein expression, and the VEGFR2 expression vector alone, XCL1-VEGFR2 wild-type and mutant fusion gene plasmids can be effectively expressed and the expression amount is equivalent.
  • Example 4 Expression of insect rod cell expression vector encoding XCL1-VEGFR2 fusion protein and protein purification
  • the cell expression used in this experiment is insect cell SF9 (Spodoptera frugiperda). This cell can be cultured in attachment or suspension. Generally, SF9 cells are cultured in suspension to express a large amount of the target protein.
  • the medium is a serum-free medium, and the Gibico medium should be used for adherent culture during the virus coating stage of this experiment.
  • the suspension culture of Yiqiao Shenzhou SIM SF medium is used.
  • Amplify the virus strain add 100 ⁇ 200 ⁇ l of P0 virus to a medium with a total cell number of 18 ⁇ 20 ⁇ 10 6 and a total volume of 15ml, and use T75 for virus amplification; collect the virus after culturing for 4 ⁇ 5 days And transferred to a sterile 15ml centrifuge tube. Centrifuge at 500 ⁇ g for 5 min, and collect the supernatant and precipitate separately
  • Virus infection Use insect cells SF9 to express the target protein, expand the cells to the appropriate cell density for infection, and add the corresponding amount of virus. After about 65 hours, the cell suspension was collected.
  • the target protein is a secreted protein, so the target protein is in the supernatant.
  • the cell suspension expressing the target protein was centrifuged at 8000 rpm/4°C for 30 min in a high-speed centrifuge to separate the cells from the supernatant, and then the supernatant was taken, and then directly subjected to in vitro affinity chromatography for protein purification.
  • Protein purification constructed by affinity chromatography using polyhistidine tag has a His-tag composed of six consecutive histidines on the N-terminus of the target protein, histidine side chain
  • the imidazole group can bind metal ions (nickel or cobalt).
  • the His-tag fusion protein is adsorbed on the column, and all the miscellaneous proteins flow out; then use a higher concentration
  • the imidazole solution (such as 200 mM) is eluted, and the imidazole will compete with the imidazole ring of His-tag, and finally the His-tag fusion protein will be eluted.
  • the histidine tag is very small and hardly affects the function, activity or structure of the target protein, so it is widely used. The main experimental steps are as follows:
  • Binding add the cell supernatant obtained after centrifugation to the nickel column, combine at 4°C, and slowly flow the supernatant through the Ni Beads to make the target protein with the His tag fully bind to the Ni Beads;
  • step 4 Concentrate the target protein solution eluted in step 3 with a 4°C centrifuge to a final volume of 500 ⁇ l.
  • Example 5 Detection of the ability of XCL1-VEGFR2 fusion protein to bind MHC-II+CD11c+CD8a+ antigen to cross-present DC cells
  • the present invention After obtaining the purified fusion protein, the present invention first verifies whether the protein obtained from insect cells can effectively bind to MHCII+CD11c+CD8a+ antigen cross-presenting DC cells.
  • mouse whole spleen as the source of CD11c+DC cells, first take mouse spleen and use 70um pore size cell mesh to grind into single cells in 1640 medium. After centrifugation at 200g for 10 minutes, discard the supernatant and use R&D company red blood cell lysate ( Catalog No. WL2000) spleen cells were treated with split red. Specifically, the lysis solution A was diluted 10 times with distilled water to prepare a working solution, and each spleen was resuspended by adding 2 mL of working solution, and placed at room temperature for 10 minutes, during which the neutralization solution B was diluted with distilled water 10 times to prepare a working solution.
  • R&D company red blood cell lysate Catalog No. WL2000
  • the cells and the magnetic bead mixture were transferred to the magnetic bead adsorption column together, and the column was placed on the magnetic stand. After the cells were completely inserted into the column, the cells were washed three times with 3 mL of 1% inactivated FBS PBS buffer. Afterwards, the column was removed from the magnetic stand and placed on the top of the 15 mL centrifuge tube. 5 mL of 1% FBS-inactivated PBS buffer was added and quickly pushed to elute the CD11c-positive DC cells, counted and centrifuged at 200 g for 10 min. The finally obtained CD11c-positive DC cells were resuspended to 1*10 6 per mL with serum-free 1640.
  • the cells were divided into BSA protein control group, VEGFR2 protein control group and XCL1-VEGFR2 wild-type protein experimental group.
  • the total amount of protein in each group was 50 ⁇ g, mixed well, and incubated in a 37° carbon dioxide incubator for 40 minutes.
  • the cells were collected and centrifuged at 500g. , And washed twice with 1% inactivated FBS PBS buffer. And perform flow dyeing: MHC-II APC, Flag-dye light 488, CD8 ⁇ -Percp/Cy5.5.
  • Use BD LSRII instrument to collect cells at medium speed, and analyze the ratio of MHC-II+Flag-dye light 488+CD8 ⁇ + cells between different groups. Repeat 3 times for each group.
  • 700 ⁇ L of the control medium containing 70ng of BSA protein, 70ng of VEGFR2 protein, and 70ng of XCL1-VEGFR2 wild-type and mutant proteins of the experimental group were added to ensure that the liquid level of the upper and lower media was consistent and prevent the upper cells from The liquid level is different and enters the lower layer due to pressure.
  • Use BD Canto II instrument to collect cells at medium speed, and analyze the ratio of MHC-II+CD11c-PE+CD8 ⁇ + cells between different groups. Repeat 3 times for each group.
  • Example 7 Intervention effect of fusion gene on the occurrence of B16 melanoma cell allograft tumor
  • the fusion protein can also effectively chemoattract and combine with MHC-II+CD11c+CD8a+ antigen to cross-present DC cells.
  • B16 melanoma cell culture and detection of VEGFR2 gene expression after tumorigenesis Resuscitate B16 cells: pre-warm cell culture medium (cell culture medium is prepared by adding 10% FBS to Corning's DMEM medium). Take out the frozen cells to be resuscitated from liquid nitrogen or -80°C refrigerator, and quickly melt them in a 37°C water bath. After the cells were completely lysed, centrifuged at 1500 rpm for 3 min, and at the same time added 8 mL of pre-warmed cell culture medium to the petri dish. After centrifugation, the supernatant was discarded, and 1 mL of pre-warmed cell culture medium was added. The cells were resuspended and transferred to a prepared petri dish.
  • the cell name, date, and number of passages were marked.
  • the cells were cultured in a 37°C, 5% CO2 incubator. Passage the next day, pre-warm the cell culture medium, trypsin, and PBS buffer. Use a vacuum pump to suck out the old medium in the petri dish. Add 7mL PBS buffer to rinse the cells once and aspirate the PBS. Add 1mL trypsin and place it in a 37°C incubator for proper time for digestion.
  • mice After confirming that the B16 cell tumorigenesis model is available, we injected the mice with gene gun plasmids according to the immunization strategy marked on the time axis in Figure 8A.
  • C57B6 purchased from Tonglihua
  • week-old male mice were divided into three groups injected with separate VEGFR2 and XCL1-VEGFR2 wild-type and mutant plasmids, five in each group, and depilatory cream was used to carry out the right side of the mouse near the groin Hair removal treatment at the lymph nodes.
  • the uppermost tumor cells are left with the middle layer of lymphocytes, washed and counted, and finally the cells are resuspended in 1640 medium containing 10% heat-inactivated FBS at a concentration of 1*10 6 cells per mL, and the cells are plated in a 24-well plate. 1 mL per well was stimulated by adding VEGFR2 epitope polypeptide, and Golgi stop (Biolegend) was added to inhibit the secretion of intracellular factors. After 12 hours, the collected cells were washed twice with 1% FBS-inactivated PBS buffer, and subjected to flow cytometry: Gran B-PE, CD8 ⁇ -Percp/Cy5.5. The results are shown in Figure 9.
  • mice immunized with XCL1-VEGFR2 wild-type and mutant plasmids have significantly more VEGFR2-specific T cells compared with the VEGFR2 immunization group alone, indicating that XCL1-VEGFR2 wild-type and mutant types Plasmid immunization did induce specific CD8a cytotoxic T lymphocytes.
  • Example 8 Therapeutic effect of T cell adoptive therapy on melanoma after immunization with fusion protein
  • the present invention uses the fusion protein purified from insect rod cells to immunize the T cell donor mouse. Induce specific cytotoxic T lymphocytes against VEGFR2 in mice, and adopt them to recipient mice with B16 tumor cells to see if it can effectively inhibit the occurrence and development of tumors in recipient mice.
  • mice were sacrificed one week after the second subcutaneous protein immunization, and the mouse spleens were taken and ground according to the method in Example 5, split red and enriched with CD8a magnetic beads for cytotoxic T lymphocytes, and finally the obtained cells were resuspended In the serum-free 1640 medium, the concentration is 2* 107 cells per mL.
  • the T cells obtained by adopting the tail vein again each had a total of 200 ⁇ L. 2*10 6 cells.
  • the mice were dissected and the lung tissues of the mice were fixed in 10% paraformaldehyde. The photos were taken the next day.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Toxicology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Provided is a VEGFR2 (KDR)-targeted metastatic cancer vaccine. By means of fusion expression of VEGFR2 protein promoting generation of tumor angiogenesis with a DC cell ligand XCL1, the efficiency of cytophagy, processing and presentation of the VEGFR2 protein by DC cells is improved, and the effect of inhibiting tumor growth is improved. Experiments prove that the protein for nucleic acid vaccine expression can effectively bind the DC cells, induce specific T-cell reaction of VEGFR2 and significantly inhibit growth of tumor highly expressing VEGFR2 in various models.

Description

靶向VEGFR2的转移性癌疫苗Metastatic cancer vaccine targeting VEGFR2
本申请要求于2020年04月09日提交中国专利局、申请号为202010273819.0、发明名称为“靶向VEGFR2的转移性癌疫苗”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。This application claims the priority of a Chinese patent application filed with the Chinese Patent Office, the application number is 202010273819.0, and the invention title is "VEGFR2-targeted metastatic cancer vaccine" on April 9, 2020, the entire content of which is incorporated into this application by reference middle.
技术领域Technical field
本发明涉及癌症的免疫治疗和预防领域,特别涉及靶向VEGFR2的转移性癌疫苗。The present invention relates to the field of cancer immunotherapy and prevention, in particular to a metastatic cancer vaccine targeting VEGFR2.
背景技术Background technique
肿瘤转移是癌症致死的主要原因,90%以上的癌症致死率是由于转移造成,而肿瘤血管的形成对于肿瘤的生长和转移则是必不可少的(Valastyan and Weinberg)。肿瘤血管为肿瘤的生长和转移提供充分的氧气和营养。而新生血管的形成则是肿瘤血管形成的必要步骤。新生血管形成理论认为肿瘤会从已存在的血管诱导新生血管的形成。基于此理论,研究者们开发出了一系列的靶向肿瘤新生血管的药物,目前FDA批准的抗血管生成药物主要分三类,单克隆抗体,受体酪氨酸激酶抑制剂以及融合的多肽。例如贝伐单抗,索拉菲尼以及VEGFR1/2蛋白胞外区的融合蛋白等用于转移性癌症的治疗,均可有效地改善病人的无进展生存期(Cascone and Heymach)。然而目前抗血管生成药物仍然存在很多问题,以贝伐单抗为例,需要频繁的肠外给药,副作用严重,明显的耐药性以及转移后肿瘤的反弹进展等,这些问题亟需解决。Tumor metastasis is the main cause of cancer death. More than 90% of cancer death rates are caused by metastasis, and the formation of tumor blood vessels is essential for tumor growth and metastasis (Valastyan and Weinberg). Tumor blood vessels provide sufficient oxygen and nutrients for tumor growth and metastasis. The formation of new blood vessels is a necessary step for tumor blood vessel formation. The neovascularization theory believes that tumors induce the formation of new blood vessels from existing blood vessels. Based on this theory, researchers have developed a series of drugs that target tumor neovascularization. The current FDA-approved anti-angiogenesis drugs are mainly divided into three categories: monoclonal antibodies, receptor tyrosine kinase inhibitors, and fusion peptides. . For example, bevacizumab, sorafenib, and the fusion protein of the extracellular domain of the VEGFR1/2 protein used in the treatment of metastatic cancer can effectively improve the patient's progression-free survival (Cascone and Heymach). However, there are still many problems with anti-angiogenic drugs. Taking bevacizumab as an example, frequent parenteral administration, serious side effects, obvious drug resistance, and tumor rebound progression after metastasis are needed. These problems need to be resolved urgently.
血管内皮生长因子(VEGF)是新生血管形成的最重要调节因子,其发挥功能主要通过结合三个受体酪氨酸激酶VEGFR1/2/3。研究表明VEGFR1高亲和VEGF但胞内酶活性却很低因而对新生血管的生成起负反馈的抑制作用,VEGFR3则主要表达在成人的淋巴管,而VEGFR2则是主要介导VEGF诱导的新生血管生成以及内皮细胞渗透性增加的促进因子(Dong et al.)。VEGFR2敲除的小鼠发育到8.5-9.5天即因为血管生成受损而夭折(Shalaby et al.)。在肿瘤组织中,VEGFR2在肿瘤组织中的肿 瘤相关内皮细胞中高表达,其表达量远高于正常血管细胞,而且,除了肿瘤相关内皮细胞,VEGFR2也在某些类型肿瘤细胞中高表达,例如B细胞淋巴瘤,乳腺癌,肺癌,膀胱癌和黑色素瘤等等(Decaussin et al.,1999;El-Obeid et al.,2004;Gakiopoulou-Givalou et al.,2003;Kranz et al.,1999),因而VEGFR2是肿瘤治疗的理想靶点。所述贝伐单抗以及索拉菲尼皆是以VEGFR2为靶点开发出的抗肿瘤药物。Vascular endothelial growth factor (VEGF) is the most important regulator of neovascularization, and its function is mainly through the binding of three receptor tyrosine kinases VEGFR1/2/3. Studies have shown that VEGFR1 has high affinity for VEGF but low intracellular enzyme activity, which has a negative feedback inhibitory effect on the formation of new blood vessels. VEGFR3 is mainly expressed in adult lymphatic vessels, while VEGFR2 is the main mediator of VEGF-induced new blood vessels. Promoting factors (Dong et al.) that produce and increase endothelial cell permeability. VEGFR2 knockout mice developed to 8.5-9.5 days and died due to impaired angiogenesis (Shalaby et al.). In tumor tissues, VEGFR2 is highly expressed in tumor-associated endothelial cells in tumor tissues, and its expression is much higher than that of normal vascular cells. In addition to tumor-associated endothelial cells, VEGFR2 is also highly expressed in certain types of tumor cells, such as B cells Lymphoma, breast cancer, lung cancer, bladder cancer and melanoma, etc. (Decaussin et al., 1999; El-Obeid et al., 2004; Gakiopoulou-Givalou et al., 2003; Kranz et al., 1999), thus VEGFR2 is an ideal target for tumor therapy. The bevacizumab and sorafenib are anti-tumor drugs developed with VEGFR2 as the target.
血管内皮生长因子受体2(VEGFR2)虽然在某些肿瘤以及肿瘤相关内皮细胞高表达,但是由于VEGFR2是一种胚胎性肿瘤相关抗原,在机体细胞免疫发育过程中,能与之识别并以高亲和力结合的T细胞在胸腺内通过阴性选择已被清除(Houghton et al.,2007)。只有对VEGFR2亲和力较低的T细胞在胸腺的选择后存活,然而这些T细胞无法被机体自身的VEGFR2所激活。因而虽然肿瘤细胞高表达VEGFR2但是机体自身并不能产生有效识别VEGFR2高表达的肿瘤细胞的效应T细胞进而抑制血管生成以及肿瘤生长。这种现象叫做免疫耐受。而既往研究表明通过将VEGFR2这类肿瘤相关抗原中的某些表位进行与T细胞的亲和力增强的突变优化,可以促进原本不识别VEGFR2这类肿瘤相关抗原的T细胞的产生,因而打破VEGFR2的免疫耐受性(Guevara-Patino et al.,2006)。Although vascular endothelial growth factor receptor 2 (VEGFR2) is highly expressed in some tumors and tumor-associated endothelial cells, because VEGFR2 is an embryonic tumor-associated antigen, it can be recognized and highly expressed during the body’s cellular immune development. Affinity-bound T cells have been eliminated by negative selection in the thymus (Houghton et al., 2007). Only T cells with low affinity for VEGFR2 survive the selection of the thymus, but these T cells cannot be activated by the body's own VEGFR2. Therefore, although tumor cells highly express VEGFR2, the body itself cannot produce effector T cells that can effectively recognize tumor cells with high expression of VEGFR2, thereby inhibiting angiogenesis and tumor growth. This phenomenon is called immune tolerance. Previous studies have shown that by optimizing certain epitopes in tumor-associated antigens such as VEGFR2 to increase their affinity with T cells, it can promote the production of T cells that do not recognize tumor-associated antigens such as VEGFR2, thereby breaking the VEGFR2 Immune tolerance (Guevara-Patino et al., 2006).
既往的研究表明,使用预测并验证的HLA-A2和HLA-A24限制性VEGFR2免疫源性多肽(分别为VEGFR2169-177:RFVPDGNRI和VEGFR2773-781:VLAMFFWLL)免疫晚期转移性结直肠癌患者以及神经纤维瘤患者,进行I/II期临床试验,结果显示,经多肽疫苗刺激后,VEGFR2特异性细胞毒性T细胞(CTLs)比例增高能抑制肿瘤新生血管生成,增加瘤内CD8+T细胞数量,并消除VEGFR2阳性的肿瘤细胞,降低肿瘤负荷,提高患者整体生存率(Hazama et al.,2014)和(Tamura et al.,2019)。但是他们的研究内容只适用于携带有HLA-A2和A24这两种亚型的人群,由于T细胞免疫应答存在HLA限制性并且多肽免疫一般不能有效引起抗体介导的体液免疫反应,所以HLA-A2和HLA-A24限制性VEGFR免疫源性多肽可能对其他HLA亚型人群无任何作用。且所用免疫多肽未经表位优化所能引起的免疫反应程度有限,这可能限制了这类疫苗的有效程度。综 合上述,有必要设计一种靶向VEGFR2的能够有效打破免疫耐受,通过细胞特异性免疫反应拮抗新生血管生成并抑制转移性癌灶形成的预防以及治疗性疫苗。Previous studies have shown that the predicted and verified HLA-A2 and HLA-A24-restricted VEGFR2 immunogenic peptides (VEGFR2169-177: RFVPDGNRI and VEGFR2773-781: VLAMFFWLL, respectively) are used to immunize patients with advanced metastatic colorectal cancer and nerve fibers Tumor patients, conducted phase I/II clinical trials, the results showed that after stimulation with peptide vaccines, the increased proportion of VEGFR2-specific cytotoxic T cells (CTLs) can inhibit tumor angiogenesis, increase the number of CD8+ T cells in the tumor, and eliminate VEGFR2-positive tumor cells reduce tumor burden and improve the overall survival rate of patients (Hazama et al., 2014) and (Tamura et al., 2019). However, their research content is only applicable to people with the two subtypes of HLA-A2 and A24. Due to the HLA-restricted T cell immune response and polypeptide immunity generally cannot effectively cause antibody-mediated humoral immune responses, HLA- A2 and HLA-A24 restricted VEGFR immunogenic peptides may not have any effect on other HLA subtypes. Moreover, the immunological peptides used without epitope optimization can cause a limited degree of immune response, which may limit the effectiveness of this type of vaccine. In summary, it is necessary to design a preventive and therapeutic vaccine that targets VEGFR2 that can effectively break immune tolerance, antagonize angiogenesis and inhibit the formation of metastatic cancer foci through cell-specific immune responses.
抗原提呈细胞指的是可以加工抗原并以抗原肽-MHC分子复合物的形式将抗原肽提呈给T细胞的一类细胞,在机体的免疫识别、免疫应答以及免疫调节中起重要作用。树突状细胞(DC)细胞是机体抗原提呈细胞的一种,并且是机体内唯一一种可以将抗原肽呈递给初始T细胞并诱导T细胞活化增殖的、功能最强的抗原呈递细胞,是机体适应性免疫应答的始动者。DC细胞根据形态以及功能可以分为经典DC以及浆细胞样DC两大类。浆细胞样DC主要介导病毒感染后的适应性免疫应答,经典DC加工呈递抗原的能力最强,则介导机体内死亡细胞的清除以及肿瘤细胞的识别等过程(Carreno et al.,2017;Minetto et al.,2019)。因而将肿瘤抗原更有效的呈递给经典DC是疫苗成功的关键,研究发现经典DC表达一类趋化因子XCL1的受体,因而XCL1分子可以高效的结合经典DC细胞(Dorner et al.,2009)。Antigen presenting cells refer to a type of cells that can process antigens and present antigen peptides to T cells in the form of antigen peptide-MHC molecular complexes, and play an important role in the body's immune recognition, immune response and immune regulation. Dendritic cell (DC) cell is a type of antigen-presenting cell in the body, and it is the only antigen-presenting cell in the body that can present antigen peptides to naive T cells and induce T cell activation and proliferation. , Is the initiator of the body's adaptive immune response. According to morphology and function, DC cells can be divided into two categories: classic DC and plasma cell-like DC. Plasma cell-like DCs mainly mediate the adaptive immune response after virus infection. Classical DCs have the strongest ability to process and present antigens, which mediate the elimination of dead cells in the body and the recognition of tumor cells (Carreno et al., 2017; Minetto et al., 2019). Therefore, more effective presentation of tumor antigens to classic DCs is the key to the success of the vaccine. Studies have found that classic DCs express a class of chemokine XCL1 receptors, so XCL1 molecules can efficiently bind to classic DC cells (Dorner et al., 2009) .
发明内容Summary of the invention
有鉴于此,本发明提供了靶向VEGFR2的转移性癌疫苗。本发明通过将促进肿瘤新生血管生成的VEGFR2蛋白与DC细胞配体XCL1融合表达,提高了VEGFR2蛋白被DC细胞吞噬、加工、呈递的效率,改善了其抑制肿瘤生长的效果。经由实验验证,本发明的核酸疫苗表达的蛋白可以有效地结合DC细胞、诱导VEGFR2特异性T细胞反应并在多种模型中显著抑制高表达VEGFR2的黑色素瘤的生长。In view of this, the present invention provides a metastatic cancer vaccine targeting VEGFR2. The invention improves the efficiency of phagocytosis, processing and presentation of the VEGFR2 protein by DC cells by fusion expression of VEGFR2 protein which promotes tumor angiogenesis and DC cell ligand XCL1, and improves the effect of inhibiting tumor growth. Experiments have shown that the protein expressed by the nucleic acid vaccine of the present invention can effectively bind to DC cells, induce VEGFR2-specific T cell responses, and significantly inhibit the growth of melanomas that highly express VEGFR2 in a variety of models.
为了实现上述发明目的,本发明提供以下技术方案:In order to achieve the above-mentioned purpose of the invention, the present invention provides the following technical solutions:
本发明提供的融合蛋白包括促进肿瘤新生血管生成的VEFGR2的能够诱发特异性CD8+T反应的胞外区或其MHCI类分子结合表位优化形式胞外区;接头;以及特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1蛋白;The fusion protein provided by the present invention includes the extracellular region of VEFGR2 that promotes tumor angiogenesis that can induce a specific CD8+T response or the extracellular region of the optimized form of binding epitope of MHC class I molecules; linkers; and specific binding with antigen crossover Human or mouse XCL1 protein of DC cells that present the ability;
其中:in:
(Ⅰ)、所述促进肿瘤新生血管生成的VEFGR2的能够诱发特异性CD8+T反应的胞外区或其MHCI类分子结合表位优化形式胞外区具有如SEQ ID No.1或2中第20~764位氨基酸所示的氨基酸序列;所述特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1,如SEQ ID NO:3或4的1~114位氨基酸所示序列;(I) The extracellular region of VEFGR2 that promotes tumor angiogenesis that can induce a specific CD8+T response or its MHC class I molecule binding epitope optimized form has the extracellular region as shown in SEQ ID No. 1 or 2. The amino acid sequence shown in amino acids 20 to 764; the human or mouse XCL1 that specifically binds to DC cells with antigen cross-presenting ability, as shown in the sequence of amino acids 1 to 114 in SEQ ID NO: 3 or 4;
or
(Ⅱ)、如(Ⅰ)所述的氨基酸序列经取代、缺失或添加一个或两个氨基酸残基获得的氨基酸序列,且与(Ⅰ)所示的氨基酸序列功能相同或相似的氨基酸序列;(II) An amino acid sequence obtained by substituting, deleting or adding one or two amino acid residues to the amino acid sequence described in (I), and having the same or similar function as the amino acid sequence shown in (I);
or
(III)、与(Ⅰ)或(Ⅱ)所述序列具有至少90%序列一致性,且与(Ⅰ)所示的氨基酸序列功能相同或相似的氨基酸序列。(III) An amino acid sequence that has at least 90% sequence identity with the sequence described in (I) or (II), and is functionally identical or similar to the amino acid sequence shown in (I).
在本发明的一些具体实施方案中,所述融合蛋白包括SEQ如ID No.7和8中的1~889位氨基酸所示序列。In some specific embodiments of the present invention, the fusion protein includes the sequence shown in amino acids 1 to 889 in SEQ ID Nos. 7 and 8.
在上述研究的基础上,本发明还提供了编码所述的融合蛋白的核苷酸,具有On the basis of the above research, the present invention also provides a nucleotide encoding the fusion protein, which has
(Ⅰ)、如SEQ ID No.11~16任意所示的核苷酸序列;(Ⅰ) The nucleotide sequence shown in SEQ ID No. 11-16;
or
(Ⅱ)、如SEQ ID No.11~16任意所示的核苷酸序列的互补核苷酸序列;或(II) The complementary nucleotide sequence of the nucleotide sequence arbitrarily shown in SEQ ID No. 11-16; or
(Ⅲ)、与(Ⅰ)或(Ⅱ)的核苷酸序列编码相同蛋白质,但因遗传密码的简并性而与(Ⅰ)或(Ⅱ)的核苷酸序列不同的核苷酸序列;(Ⅲ). A nucleotide sequence that encodes the same protein as the nucleotide sequence of (I) or (II), but is different from the nucleotide sequence of (I) or (II) due to the degeneracy of the genetic code;
or
(Ⅳ)、与(Ⅰ)、(Ⅱ)或(Ⅲ)所示的核苷酸序列经取代、缺失或添加一个或两个核苷酸序列获得的核苷酸序列,且与(Ⅰ)、(Ⅱ)或(Ⅲ)所示的核苷酸序列功能相同或相似的核苷酸序列;(IV), the nucleotide sequence obtained by substituting, deleting or adding one or two nucleotide sequences to the nucleotide sequence shown in (I), (II) or (III), and the nucleotide sequence with (I), The nucleotide sequence shown in (II) or (III) has the same or similar function;
or
(V)、与(Ⅰ)、(Ⅱ)、(Ⅲ)或(Ⅳ)所述核苷酸序列具有至少90%序列一致性的核苷酸序列。(V), a nucleotide sequence that has at least 90% sequence identity with the nucleotide sequence described in (I), (II), (III) or (IV).
在上述研究的基础上,本发明还提供了重组表达载体,包括载体以及本发明所述的融合蛋白或所述的核苷酸。On the basis of the above research, the present invention also provides a recombinant expression vector, including a vector and the fusion protein or the nucleotide of the present invention.
在本发明的一些具体实施方案中,所述载体包括哺乳动物细胞表达载体或昆虫杆状细胞表达载体;所述载体包括pcDNA3.1(+)、pcDNA3.1(-)、pFastbac1-dual-MBP。In some specific embodiments of the present invention, the vector includes a mammalian cell expression vector or an insect rod-shaped cell expression vector; the vector includes pcDNA3.1(+), pcDNA3.1(-), pFastbac1-dual-MBP .
在上述研究的基础上,本发明还提供了包括本发明所述的融合蛋白或所述的核苷酸的重组菌株或细胞。On the basis of the above research, the present invention also provides a recombinant strain or cell comprising the fusion protein or the nucleotide of the present invention.
在上述研究的基础上,本发明还提供了所述融合蛋白、所述核苷酸、所述重组表达载体或所述的重组菌株或细胞在制备转移性癌症或VEGFR2高表达的黑色素瘤的疫苗或制备预防和/或治疗转移性癌症或VEGFR2高表达的黑色素瘤的药物中的应用。On the basis of the above research, the present invention also provides a vaccine for preparing metastatic cancer or melanoma with high expression of VEGFR2 in the fusion protein, the nucleotide, the recombinant expression vector or the recombinant strain or cell. Or application in the preparation of medicines for preventing and/or treating metastatic cancer or melanoma with high expression of VEGFR2.
在本发明的一些具体实施方案中,所述转移性癌症包括肝癌,结直肠癌或肺癌。In some specific embodiments of the present invention, the metastatic cancer includes liver cancer, colorectal cancer or lung cancer.
在上述研究的基础上,本发明还提供了转移性癌症或VEGFR2高表达的黑色素瘤的疫苗,包括本发明所述的融合蛋白、所述的核苷酸、所述的重组表达载体以及药学上可接受的运载体、赋形剂和/或佐剂。On the basis of the above research, the present invention also provides a vaccine for metastatic cancer or melanoma with high expression of VEGFR2, including the fusion protein of the present invention, the nucleotide, the recombinant expression vector and the pharmaceutical Acceptable carriers, excipients and/or adjuvants.
在上述研究的基础上,本发明还提供了预防和/或治疗转移性癌症或VEGFR2高表达的黑色素瘤的药物,包括本发明所述的融合蛋白、所述的核苷酸、所述的重组表达载体以及药学上可接受的辅料。On the basis of the above research, the present invention also provides drugs for the prevention and/or treatment of metastatic cancer or melanoma with high expression of VEGFR2, including the fusion protein of the present invention, the nucleotide, and the recombinant Expression vector and pharmaceutically acceptable excipients.
本发明提供的疫苗是由血管内皮细胞生长因子受体VEGFR2的能够引起特异性CD8+T细胞免疫应答的胞外区以及MHCI类分子结合表位优化形式的胞外区并在其N端融合表达特异结合抗原交叉呈递能力的DC的XCL1分子构成的蛋白,包含所述融合蛋白的核酸以及核酸所在的载体,包含本发明所述融合蛋白,核酸和载体在预防和治疗黑色素瘤,转移性癌中的应用。本发明通过将促进肿瘤新生血管生成的VEGFR2蛋白与DC细胞配体XCL1融合表达,提高了VEGFR2蛋白被DC细胞吞噬、加工、呈递的效率,改善了其抑制肿瘤生长的效果。经由实验验证,本发明的核酸疫苗表达的蛋白可以有效地结合DC细胞、诱导VEGFR2特异性T细胞反应并在多种模型中显著抑制高表达VEGFR2的黑色素瘤的生长。The vaccine provided by the present invention is composed of the extracellular region of the vascular endothelial cell growth factor receptor VEGFR2 that can cause a specific CD8+ T cell immune response and the extracellular region of the optimized form of MHC class I molecule binding epitope, and is expressed by fusion at its N-terminus The protein composed of the XCL1 molecule of DC that specifically binds to the antigen cross-presenting ability, contains the nucleic acid of the fusion protein and the carrier where the nucleic acid is located, and contains the fusion protein of the present invention. The nucleic acid and the carrier are used in the prevention and treatment of melanoma and metastatic cancer. Applications. The invention improves the efficiency of phagocytosis, processing and presentation of the VEGFR2 protein by DC cells by fusion expression of VEGFR2 protein which promotes tumor angiogenesis and DC cell ligand XCL1, and improves the effect of inhibiting tumor growth. Experiments have shown that the protein expressed by the nucleic acid vaccine of the present invention can effectively bind to DC cells, induce VEGFR2-specific T cell responses, and significantly inhibit the growth of melanomas that highly express VEGFR2 in a variety of models.
附图说明Description of the drawings
为了更清楚地说明本发明实施例或现有技术中的技术方案,下面将对实施例或现有技术描述中所需要使用的附图作简单地介绍。In order to explain the embodiments of the present invention or the technical solutions in the prior art more clearly, the following will briefly introduce the drawings that need to be used in the description of the embodiments or the prior art.
图1示编码融合蛋白核苷酸的载体图谱;其中图1A示CMV启动子之后接XCL1序列之后连接VEGFR2野生型胞外区用于核酸疫苗编码序列;图1B示CMV启动子之后接XCL1序列之后连接VEGFR2MHCI类分子结合优化突变型胞外区核酸疫苗编码序列;图1C示CMV启动子之后接XCL1分泌信号肽序列之后连接VEGFR2胞外区用于核酸疫苗编码序列;图1D示pFASTbac-dual-MBP-XCL1-VEGFR2野生型融合蛋白疫苗纯化载体;图1E示pFASTbac-dual-MBP-XCL1-VEGFR2突变型融合蛋白疫苗纯化载体;图1F示pFASTbac-dual-MBP-VEGFR2野生型蛋白疫苗纯化载体;Figure 1 shows the vector map of the nucleotide encoding the fusion protein; Figure 1A shows the CMV promoter followed by the XCL1 sequence followed by the VEGFR2 wild-type extracellular region for the nucleic acid vaccine coding sequence; Figure 1B shows the CMV promoter followed by the XCL1 sequence Connecting VEGFR2MHCI molecules to optimize the mutant extracellular region nucleic acid vaccine coding sequence; Figure 1C shows the CMV promoter followed by the XCL1 secretion signal peptide sequence followed by the VEGFR2 extracellular region for nucleic acid vaccine coding sequence; Figure 1D shows pFASTbac-dual-MBP -XCL1-VEGFR2 wild-type fusion protein vaccine purification vector; Figure 1E shows the pFASTbac-dual-MBP-XCL1-VEGFR2 mutant fusion protein vaccine purification vector; Figure 1F shows the pFASTbac-dual-MBP-VEGFR2 wild-type protein vaccine purification vector;
图2示融合蛋白的三维结构预测结果图;将编码融合蛋白的核苷酸序列输入http://raptorx.uchicago.edu/网址对融合蛋白的三维空间结构预测,图示XCL1-VEGFR2融合蛋白空间结构图;Figure 2 shows the results of the three-dimensional structure prediction of the fusion protein; input the nucleotide sequence encoding the fusion protein into the website http://raptorx.uchicago.edu/ to predict the three-dimensional structure of the fusion protein, showing the XCL1-VEGFR2 fusion protein space Structure diagram
图3示检测编码融合蛋白的核酸在HEK293T细胞内的表达情况;将载由编码融合蛋白核酸的质粒载体转染HEK292T细胞48小时后,利用蛋白免疫印迹技术(Westernblot)检测C端带有Flag标签的融合蛋白编码核苷酸的表达情况;Figure 3 shows the detection of the expression of the nucleic acid encoding the fusion protein in HEK293T cells; after 48 hours of transfection of the plasmid vector carrying the nucleic acid encoding the fusion protein into HEK292T cells, Western blotting was used to detect the C-terminal with a Flag tag The expression of nucleotides encoding the fusion protein;
图4示检测从昆虫杆状细胞SF9中纯化的融合蛋白;利用考马斯亮蓝染色检测从昆虫杆状细胞分泌表达的XCL1-VEGFR2的纯化情况;Figure 4 shows the detection of the fusion protein purified from insect rod-shaped cells SF9; Coomassie brilliant blue staining is used to detect the purification of XCL1-VEGFR2 secreted and expressed from insect rod-shaped cells;
图5示融合蛋白可以有效地结合MHCII+CD11c+CD8a+抗原交叉呈递DC细胞;将带有Flag标签的SF9细胞纯化的蛋白与CD11C磁珠富集的脾细胞孵育,利用Flag荧光抗体检测融合蛋白与MHCII+CD11c+CD8a+抗原交叉呈递DC细胞的结合情况;其中,图5(A)示融合蛋白结合DC细胞流式分析图;图5(B)示5(A)中融合蛋白结合DC细胞比例统计分析图;Figure 5 shows that the fusion protein can effectively bind to MHCII+CD11c+CD8a+ antigen cross-presenting DC cells; the Flag-tagged SF9 cell-purified protein is incubated with the spleen cells enriched with CD11C magnetic beads, and Flag fluorescent antibody is used to detect the fusion protein and MHCII+CD11c+CD8a+ antigen cross-presenting DC cell binding situation; among them, Figure 5 (A) shows the fusion protein binding DC cell flow analysis diagram; Figure 5 (B) shows the statistics of the proportion of fusion protein binding DC cells in 5 (A) diagram;
图6示检测融合蛋白对MHCII+CD11c+CD8a+抗原交叉呈递DC细胞的趋化效应;利用Transwell试验,将含有MHCII+CD11c+CD8a+抗原交叉呈递DC细胞的脾细胞放在上层,将从SF9昆虫细胞中纯化的融合蛋白放于下层,观察2小时后上层MHCII+CD11c+CD8a+抗原交叉呈递DC细胞进 入下层的数量;结果显示融合蛋白对MHCII+CD11c+CD8a+抗原交叉呈递DC细胞具有趋化作用;其中,图6(A)示融合蛋白趋化DC细胞流式分析图;图6(B)示6(A)融合蛋白趋化DC细胞比例统计分析图;Figure 6 shows the detection of the chemotactic effect of the fusion protein on MHCII+CD11c+CD8a+ antigen cross-presenting DC cells; using the Transwell test, spleen cells containing MHCII+CD11c+CD8a+ antigen cross-presenting DC cells are placed on the upper layer, and SF9 insect cells will be removed Place the purified fusion protein in the lower layer and observe the number of upper MHCII+CD11c+CD8a+ antigen cross-presenting DC cells entering the lower layer after 2 hours; the results show that the fusion protein has a chemotactic effect on MHCII+CD11c+CD8a+ antigen cross-presenting DC cells; , Figure 6(A) shows a flow cytometric analysis chart of fusion protein chemotactic DC cells; Figure 6(B) shows a statistical analysis chart of the proportion of 6(A) fusion protein chemotactic DC cells;
图7示检测黑色素瘤细胞系B16细胞成瘤后的VEGFR2(KDR)表达情况;将肿瘤细胞接种于皮下,待成瘤后,取肿瘤组织以及周边皮肤组织提取总RNA,利用实时荧光定量PCR扩增技术检测VEGFR2在肿瘤以及周边皮肤的表达情况,结果显示VEGFR2在肿瘤组织中的表达显著高于周边正常皮肤;Figure 7 shows the detection of VEGFR2 (KDR) expression of melanoma cell line B16 cells after tumorigenesis; tumor cells are inoculated subcutaneously, after tumorigenesis, the tumor tissue and surrounding skin tissue are taken to extract total RNA, and real-time fluorescent quantitative PCR is used to expand The augmentation technique detects the expression of VEGFR2 in the tumor and surrounding skin, and the results show that the expression of VEGFR2 in the tumor tissue is significantly higher than that in the surrounding normal skin;
图8示编码XCL1-VEGFR2/XCL1-VEGFR2突变型融合蛋白的融合基因免疫小鼠后,小鼠黑色素瘤的生长情况;图8A示融合基因免疫方法以及免疫过程图;图8B示接种黑色素瘤后,测量肿瘤的体积做出的肿瘤生长曲线图;图8C示待对照组肿瘤生长至2000mm 3的伦理终点时处死小鼠,解剖出肿瘤的大小比较图; Figure 8 shows the growth of mouse melanoma after immunization with the fusion gene encoding XCL1-VEGFR2/XCL1-VEGFR2 mutant fusion protein; Figure 8A shows the fusion gene immunization method and the immunization process diagram; Figure 8B shows the melanoma after inoculation , A graph of tumor growth curve made by measuring the volume of the tumor; Figure 8C shows that the mice were sacrificed when the tumor in the control group grew to the ethical end point of 2000 mm 3 and the tumor size comparison diagram was dissected;
图9示编码XCL1-VEGFR2融合蛋白的的融合基因免疫小鼠后,小鼠黑色素瘤内部侵润的淋巴细胞颗粒酶B阳性的CD8aT细胞所占比例分析图;Figure 9 shows an analysis diagram of the proportion of granzyme B-positive CD8a T cells in lymphocytes infiltrating the mouse melanoma after immunization with a fusion gene encoding XCL1-VEGFR2 fusion protein;
图10示利用融合蛋白免疫小鼠后,取脾脏细胞过继到静脉注射黑色素瘤细胞的小鼠体内,检测肺部模拟黑色素瘤转移结节灶的形成情况;图10A示融合蛋白用静脉注射免疫的方式效果图;图10B示融合蛋白免疫过继治疗后小鼠肺部结节图。Figure 10 shows the use of the fusion protein to immunize mice, and then the spleen cells are taken into the mice that were intravenously injected with melanoma cells to detect the formation of simulated melanoma metastatic nodules in the lungs; Figure 10A shows the fusion protein immunized by intravenous injection Method effect diagram; Figure 10B shows a diagram of mouse lung nodules after adoptive treatment with fusion protein immunization.
具体实施方式Detailed ways
本发明公开了靶向VEGFR2的转移性癌疫苗,本领域技术人员可以借鉴本文内容,适当改进工艺参数实现。特别需要指出的是,所有类似的替换和改动对本领域技术人员来说是显而易见的,它们都被视为包括在本发明。本发明的方法及应用已经通过较佳实施例进行了描述,相关人员明显能在不脱离本发明内容、精神和范围内对本文所述的方法和应用进行改动或适当变更与组合,来实现和应用本发明技术。The present invention discloses a metastatic cancer vaccine targeting VEGFR2. Those skilled in the art can learn from the content of this article and appropriately improve the process parameters to achieve it. In particular, it should be pointed out that all similar replacements and modifications are obvious to those skilled in the art, and they are all deemed to be included in the present invention. The method and application of the present invention have been described through the preferred embodiments. It is obvious that relevant persons can make changes or appropriate changes and combinations to the methods and applications described herein without departing from the content, spirit and scope of the present invention to achieve and Apply the technology of the present invention.
本发明采用了VEGFR2的胞外区的氨基酸序列作为免疫原,以方便 可溶性蛋白疫苗的纯化,本发明的疫苗可在人或小鼠体内有效诱导出针对VEGFR2的特异性T细胞免疫反应,打破了原肿瘤相关抗原VEGFR2的免疫耐受性,因而可以有效的发挥直接清除表达VEGFR2的肿瘤细胞,间接通过抑制肿瘤新生血管生成而抑制肿瘤生长以及转移灶的形成。The present invention uses the amino acid sequence of the extracellular region of VEGFR2 as an immunogen to facilitate the purification of soluble protein vaccines. The vaccine of the present invention can effectively induce specific T cell immune responses against VEGFR2 in humans or mice, breaking The immune tolerance of the original tumor-associated antigen VEGFR2 can effectively directly eliminate tumor cells expressing VEGFR2, and indirectly inhibit tumor growth and the formation of metastases by inhibiting tumor angiogenesis.
本发明一方面涉及一种转移性癌症的融合蛋白疫苗,所述疫苗由1)促进肿瘤新生血管生成的血管内皮生长因子受体2(VEFGR2)的能够诱发特异性CD8+T反应的胞外区以及其MHCI类分子结合表位优化形式胞外区,以及2)N端通过接头融合表达特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1蛋白组成。例如小鼠VEGFR2,SEQ ID No.1和SEQ ID No.2中的20-764位氨基酸所示的序列构成,其N端通过接头融合表达特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1,由SEQ ID No.3或SEQ ID No.4的1-114位氨基酸所示序列构成。One aspect of the present invention relates to a fusion protein vaccine for metastatic cancer. The vaccine consists of 1) the extracellular region of vascular endothelial growth factor receptor 2 (VEFGR2) that promotes tumor angiogenesis and can induce a specific CD8+T response And its MHC class I molecule binding epitope optimized form extracellular region, and 2) N-terminal fusion expresses the human or mouse XCL1 protein composition that specifically binds to DC cells with antigen cross-presentation ability. For example, mouse VEGFR2, SEQ ID No. 1 and SEQ ID No. 2 are composed of amino acids 20-764, and the N-terminus is expressed through a linker fusion that specifically binds human or human DC cells with antigen cross-presentation ability. Mouse XCL1 is composed of the sequence shown in amino acids 1-114 of SEQ ID No. 3 or SEQ ID No. 4.
在一些实施例中,所述转移性癌的疫苗包含SEQ ID No.7和SEQ ID No.8中的1-889位氨基酸的任意序列。In some embodiments, the vaccine for metastatic cancer includes any sequence of amino acids 1-889 in SEQ ID No. 7 and SEQ ID No. 8.
本发明另一方面涉及一种编码该融合蛋白疫苗的核酸,核酸疫苗由1)编码促进肿瘤新生血管生成的血管内皮生长因子受体2(VEFGR2)的能够诱发特异性CD8+T反应的胞外区以及其MHCI类分子结合表位优化形式胞外区的核苷酸序列,以及2)编码N端通过接头融合表达特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1蛋白的核苷酸序列组成。即编码SEQ ID No.1和SEQ ID No.2中20-764位氨基酸序列和特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1序列,即SEQ ID No.3或SEQ ID No.4中1-114位氨基酸序列。Another aspect of the present invention relates to a nucleic acid encoding the fusion protein vaccine. The nucleic acid vaccine consists of 1) encoding the extracellular vascular endothelial growth factor receptor 2 (VEFGR2) that promotes tumor angiogenesis and can induce a specific CD8+T response. Region and the nucleotide sequence of the extracellular region of the optimized form of binding epitope of MHC class I molecules, and 2) the N-terminal fusion expressing the nucleoside of human or mouse XCL1 protein that specifically binds to DC cells with antigen cross-presenting ability through linker fusion Acid sequence composition. That is, it encodes the 20-764 amino acid sequence of SEQ ID No. 1 and SEQ ID No. 2 and the human or mouse XCL1 sequence that specifically binds to DC cells with antigen cross-presentation ability, that is, SEQ ID No. 3 or SEQ ID No. 4 in the amino acid sequence of positions 1-114.
在一些实施例中,编码促进肿瘤新生血管生成的VEFGR2和特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1的序列包含SEQ ID No.11-SEQ ID No.16中的核苷酸序列。In some embodiments, the sequence encoding VEFGR2 that promotes tumor angiogenesis and human or murine XCL1 that specifically binds to DC cells with antigen cross-presenting ability includes the nucleotides in SEQ ID No. 11-SEQ ID No. 16 sequence.
本发明同时还提供了一种负载所述核酸疫苗的重组表达载体,其包含本发明所述融合蛋白疫苗以及对应核算疫苗的核苷酸序列以及载体。所述载体可以为哺乳动物细胞细胞表达载体或昆虫杆状细胞表达载体,具体的,所述载体可以为pcDNA3.1(+)、pcDNA3.1(-)、pFastbac1-dual-MBP。The present invention also provides a recombinant expression vector carrying the nucleic acid vaccine, which contains the fusion protein vaccine of the present invention and the nucleotide sequence and vector corresponding to the vaccine vaccine. The vector can be a mammalian cell expression vector or an insect rod-shaped cell expression vector. Specifically, the vector can be pcDNA3.1(+), pcDNA3.1(-), pFastbac1-dual-MBP.
本发明最终提供一种转移性癌症的疫苗,包含所述的融合蛋白疫苗,核酸疫苗以及重组表达载体。The present invention finally provides a vaccine for metastatic cancer, including the fusion protein vaccine, nucleic acid vaccine and recombinant expression vector.
本发明的疫苗为肝癌,结直肠癌,肺癌等转移性癌术后以及高表达VEGFR2的黑色素瘤提供一种预防复发或治疗的办法。包括给与其他适应症本发明的疫苗。The vaccine of the present invention provides a method for preventing recurrence or treatment for postoperative metastatic cancers such as liver cancer, colorectal cancer, lung cancer, and melanoma with high expression of VEGFR2. Including the administration of the vaccine of the present invention for other indications.
本发明的也提供了所述的疫苗,核酸疫苗以及载体在制备肝癌,结直肠癌,肺癌等转移性癌术后以及高表达VEGFR2的黑色素瘤疫苗中的应用。The present invention also provides the application of the vaccine, nucleic acid vaccine and vector in preparing liver cancer, colorectal cancer, lung cancer and other metastatic cancers after surgery and melanoma vaccines with high expression of VEGFR2.
本发明也提供了所述的疫苗,核酸疫苗以及载体用于治疗黑色素瘤。The present invention also provides the vaccine, nucleic acid vaccine and carrier for treating melanoma.
本发明的疫苗或药物包括所述融合蛋白、所述核酸、所述重组表达载体以及任选存在的药物可接受的运载体、赋形剂和/或佐剂。The vaccine or drug of the present invention includes the fusion protein, the nucleic acid, the recombinant expression vector, and optionally pharmaceutically acceptable carriers, excipients and/or adjuvants.
综上,本发明提供了转移性癌的疫苗,该疫苗是由血管内皮细胞生长因子受体VEGFR2的能够引起特异性CD8+T细胞免疫应答的胞外区以及MHCI类分子结合表位优化形式的胞外区并在其N端融合表达特异结合抗原交叉呈递能力的DC的XCL1分子构成的蛋白,包含所述融合蛋白的核酸以及核酸所在的载体,包含本发明所述融合蛋白,核酸和载体在预防和治疗黑色素瘤,转移性癌中的应用。本发明通过将促进肿瘤新生血管生成的VEGFR2蛋白与DC细胞配体XCL1融合表达,提高了VEGFR2蛋白被DC细胞吞噬、加工、呈递的效率,改善了其抑制肿瘤生长的效果。经由实验验证,本发明的核酸疫苗表达的蛋白可以有效地结合DC细胞、诱导VEGFR2特异性T细胞反应并在多种模型中显著抑制高表达VEGFR2的黑色素瘤的生长。In summary, the present invention provides a vaccine for metastatic cancer, the vaccine is optimized by the extracellular region of the vascular endothelial cell growth factor receptor VEGFR2 that can cause a specific CD8+ T cell immune response and MHC class I molecule binding epitopes. The extracellular region is fused at its N-terminus to express a protein composed of XCL1 molecules that specifically bind to the antigen cross-presenting ability, the nucleic acid containing the fusion protein and the vector in which the nucleic acid is located, including the fusion protein of the present invention, the nucleic acid and the vector Application in the prevention and treatment of melanoma and metastatic cancer. The invention improves the efficiency of phagocytosis, processing and presentation of the VEGFR2 protein by DC cells by fusion expression of VEGFR2 protein which promotes tumor angiogenesis and DC cell ligand XCL1, and improves the effect of inhibiting tumor growth. Experiments have shown that the protein expressed by the nucleic acid vaccine of the present invention can effectively bind to DC cells, induce VEGFR2-specific T cell responses, and significantly inhibit the growth of melanomas that highly express VEGFR2 in a variety of models.
本发明中涉及的原料及试剂均可由市场购得。The raw materials and reagents involved in the present invention can be purchased from the market.
下面结合实施例,进一步阐述本发明:The following examples further illustrate the present invention:
实施例1 融合蛋白疫苗抗原的设计方案以及哺乳动物细胞和昆虫杆状细胞表达质粒的构建与制备Example 1 The design scheme of fusion protein vaccine antigen and the construction and preparation of mammalian cell and insect rod-shaped cell expression plasmids
(1)融合基因哺乳动物细胞表达载体的构建:(1) Construction of fusion gene mammalian cell expression vector:
根据小鼠VEGFR2(SEQ ID No.1和SEQ ID No.2)和小鼠XCL1(SEQ  ID No.4)蛋白的氨基酸序列构建融合蛋白XCL1-VEGFR2,为了促进表达融合蛋白的核酸在体内翻译出的融合蛋白可以有效的分泌到细胞外并趋化MHC-II+CD11c+CD8a+抗原交叉呈递DC细胞,我们保留了XCL1蛋白的分泌信号肽(SEQ ID No.5),同时将VEGFR2介导细胞跨膜定位的信号肽去掉(SEQ ID No.6)。最终得到的融合蛋白的氨基酸序列(SEQ ID No.7和SEQ ID No.8)。对于单独VEGFR2蛋白则将XCL1蛋白的分泌信号肽加到去除自身信号肽的蛋白N端(SEQ ID No.9和SEQ ID No.10),以保证单独VEGFR2表达载体表达出的VEGFR2蛋白也能同样的被分泌到细胞外。我们将氨基酸序列对应的核苷酸序列进行哺乳动物细胞表达偏好的密码子优化,由擎科公司进行合成,并连接到pcDNA3.1/zeo(-)以及pFastbac1-dual-MBP表达载体上(SEQ ID No.11~SEQ ID No.16)。The fusion protein XCL1-VEGFR2 was constructed according to the amino acid sequence of mouse VEGFR2 (SEQ ID No.1 and SEQ ID No.2) and mouse XCL1 (SEQ ID No.4), in order to promote the translation of nucleic acid expressing the fusion protein in vivo The fusion protein can be effectively secreted outside the cell and chemotactic MHC-II+CD11c+CD8a+ antigen cross-presenting DC cells. We retained the secretion signal peptide of XCL1 protein (SEQ ID No.5), and at the same time, VEGFR2 mediates cell cross-presentation. The signal peptide for membrane positioning is removed (SEQ ID No. 6). The final amino acid sequence of the fusion protein (SEQ ID No. 7 and SEQ ID No. 8). For a single VEGFR2 protein, add the secretion signal peptide of the XCL1 protein to the N-terminus of the protein that removes its own signal peptide (SEQ ID No. 9 and SEQ ID No. 10) to ensure that the VEGFR2 protein expressed by the single VEGFR2 expression vector can also be the same Is secreted out of the cell. We optimized the nucleotide sequence corresponding to the amino acid sequence for mammalian cell expression preference codon optimization, synthesized it by Dynatech, and ligated it to pcDNA3.1/zeo(-) and pFastbac1-dual-MBP expression vector (SEQ ID No. 11 ~ SEQ ID No. 16).
(2)融合基因哺乳动物细胞表达载体的扩增(2) Amplification of fusion gene mammalian cell expression vector
细菌的转化,取-80℃冻存的感受态细菌置于冰上融化,待接近完全融化时加入100ng质粒,轻柔混匀,冰上放置30mins。将感受态置于42℃水浴锅中热激60s,取出后立即置于冰上放置2mins。向管内加入500μL不含抗生素的LB培养基,37℃摇床内振荡培养1h。将细菌4000rpm室温离心2mins,弃去部分上清(约450μL)后重悬细菌,取适量细菌悬液涂于含有相应抗生素的培养皿上。培养皿正面朝下放置,37℃培养箱培养过夜。待克隆大小适宜后(约16h)用枪尖将克隆挑至加好抗生素的LB培养基中,37℃摇床中振荡培养至浑浊。取15mL培养至合适浓度的细菌菌液,4000rpm室温离心5mins,弃掉上清。按照北京天根公司的《质粒小提试剂盒》货号(DP103)说明书提取细菌DNA。首先加入250μL含RNA酶的细胞重悬液P1充分重悬细菌沉淀,并将沉淀转移至1.5mL EP管中。加入250μL碱性细胞裂解液P2,轻柔颠倒混匀至液体澄清。加入350μL中和液P3,颠倒混合均匀至出现絮状沉淀物。混悬液12000rpm室温离心10mins。将DNA吸附柱放入回收管中,加入500μL平衡液活化吸附膜,12000rpm离心1min后弃掉液体。将步骤4中离心获得的上清转移至DNA吸附柱中,12000rpm离心1min后弃去液体。向吸附柱内加入600μL清洗液,12000rpm 离心1min后弃去液体,重复洗涤一次。12000rpm空离回收管2mins。将收集管更换为新的1.5mL EP管,室温放置5mins晾干吸附柱。加入70μL 65℃预热的洗脱缓冲液或者高压过的纯净水,室温放置5mins充分溶解DNA,12000rpm室温离心3mins,收集液体。弃掉吸附柱后对管内质粒进行浓度测定并标记质粒名称、浓度、提取日期。For bacterial transformation, place competent bacteria frozen at -80°C on ice and thaw, add 100ng plasmid when it is almost completely thawed, mix gently, and place on ice for 30 minutes. Place the competent state in a 42°C water bath for 60 seconds, and immediately place it on ice for 2 minutes after taking it out. Add 500 μL of antibiotic-free LB medium to the tube, and shake culture in a shaker at 37°C for 1 hour. Centrifuge the bacteria at 4000 rpm for 2 minutes at room temperature, discard a portion of the supernatant (about 450 μL) and resuspend the bacteria, and apply an appropriate amount of the bacterial suspension to a petri dish containing the corresponding antibiotic. Place the petri dish face down and incubate overnight in a 37°C incubator. After the clone size is appropriate (about 16h), use a gun tip to pick the clones into antibiotic-added LB medium, and culture them in a shaker at 37°C until they become turbid. Take 15 mL of the bacterial solution cultured to a suitable concentration, centrifuge at 4000 rpm for 5 minutes at room temperature, and discard the supernatant. Bacterial DNA was extracted according to the instruction of "Plasmid Small Extraction Kit" (DP103) of Beijing Tiangen Company. First add 250μL of RNase-containing cell resuspension P1 to fully resuspend the bacterial pellet, and transfer the pellet to a 1.5mL EP tube. Add 250μL of alkaline cell lysate P2, gently invert and mix until the liquid is clear. Add 350μL of neutralization solution P3, and mix by inversion until flocculent sediment appears. The suspension was centrifuged at 12000rpm for 10mins at room temperature. Put the DNA adsorption column into the recovery tube, add 500 μL of balance solution to activate the adsorption membrane, centrifuge at 12000 rpm for 1 min, and discard the liquid. Transfer the supernatant obtained by centrifugation in step 4 to the DNA adsorption column, centrifuge at 12000 rpm for 1 min, and discard the liquid. Add 600 μL of cleaning solution to the adsorption column, centrifuge at 12000 rpm for 1 min, discard the liquid, and repeat the washing once. Leave the recovery tube at 12000rpm for 2mins. Replace the collection tube with a new 1.5mL EP tube, and leave it at room temperature for 5 minutes to dry the adsorption column. Add 70μL of 65°C preheated elution buffer or high-pressure purified water, leave it at room temperature for 5mins to fully dissolve the DNA, centrifuge at 12000rpm for 3mins at room temperature, and collect the liquid. After the adsorption column is discarded, the concentration of the plasmid in the tube is determined and the plasmid name, concentration, and extraction date are marked.
(3)融合昆虫杆状细胞表达载体的构建(3) Construction of fusion insect rod cell expression vector
1)蓝白斑筛选的基本原理:此表达系统的第一个成分是用来克隆目的基因的pFastBac菌株。基于pFastBac菌株的选择,表达基因被PH或p10启动子控制,在昆虫细胞内高水平表达表达框被Tn7的左右臂包围,包含一个庆大霉素抗性点,形成一个微型的Tn7。此表达系统的第二个成分是E.coli的DH10Bac品系。DH10Bac细胞包含带有微型-attTn7靶位点的病毒质粒和辅助质粒。只要pFastBac表达质粒转入DH10Bac细胞,就会在pFastBac菌株的微型Tn7单位和微型-attTn7的靶位点之间发生转化,从而产生重组质粒。病毒菌株(杆粒)在具有卡那抗性的质粒DH10Bac中扩增,在显色物质X-gal和诱导剂IPTG存在时,能补充染色体LacZ的缺失形成蓝斑。微型Tn7插入到微型attTn7附属位点会干扰LacZa肽的表达,所以包含重组质粒的克隆在蓝色背景下是白色的,选择白斑分析。真正的白斑克隆比较大,因此为了避免选择成假阳性,选择最大的、最独立的白斑。1) The basic principle of blue-white screening: the first component of this expression system is the pFastBac strain used to clone the target gene. Based on the selection of the pFastBac strain, the expression gene is controlled by the PH or p10 promoter, and the high-level expression expression frame in insect cells is surrounded by the left and right arms of Tn7, which contains a gentamicin resistance point, forming a miniature Tn7. The second component of this expression system is the DH10Bac strain of E. coli. DH10Bac cells contain viral plasmids with mini-attTn7 target sites and helper plasmids. As long as the pFastBac expression plasmid is transferred into DH10Bac cells, transformation will occur between the mini-Tn7 unit of the pFastBac strain and the target site of mini-attTn7, thereby producing a recombinant plasmid. The virus strain (bacmid) is amplified in the kana-resistant plasmid DH10Bac, and when the coloring substance X-gal and the inducer IPTG are present, it can supplement the lack of chromosome LacZ to form blue spots. The insertion of micro-Tn7 into the micro-attTn7 attachment site will interfere with the expression of LacZa peptide, so the clone containing the recombinant plasmid is white on the blue background, so choose the white spot analysis. The true leukoplakia clone is relatively large, so in order to avoid false positive selection, choose the largest and most independent leukoplakia.
2)蓝白斑的制备:重组质粒pFastBac HTB的构建:使用以上介绍的实验方法进行引物设计(SEQ ID No.17~20)、琼脂糖胶回收、酶切反应、连接反应、转化和质粒提取等实验,最终得到将目的基因克隆进入pFastBac HTB的重组质粒;重组质粒pFastBac HTB转化到DH10Bac感受态细胞:挑取目的菌斑(白斑):48个小时后,直到能够明确地区分蓝白斑,选择最大最独立的白斑2~3个,用5ml LB培养基(已加入50μg/ml卡那霉素,10μg/ml四环素,7μg/ml庆大霉素)于37℃、220rpm摇8~12个小时,直到菌液浑浊,之后用此菌液进行Bacmid的提取。2) Preparation of blue and white spots: construction of recombinant plasmid pFastBac HTB: use the above-mentioned experimental method for primer design (SEQ ID No. 17-20), agarose gel recovery, enzyme digestion reaction, ligation reaction, transformation and plasmid extraction, etc. The experiment finally obtained the recombinant plasmid that cloned the target gene into pFastBac HTB; the recombinant plasmid pFastBac HTB transformed into DH10Bac competent cells: Pick the target plaque (white spot): 48 hours later, until the blue and white spots can be clearly distinguished, choose the largest The most independent leukoplakia 2 to 3, use 5ml LB medium (50μg/ml kanamycin, 10μg/ml tetracycline, 7μg/ml gentamicin) shake at 37℃, 220rpm for 8-12 hours, Until the bacterial liquid is turbid, then use this bacterial liquid to extract Bacmid.
表1Table 1
Figure PCTCN2020109970-appb-000001
Figure PCTCN2020109970-appb-000001
实施例2 融合蛋白三维结构预测Example 2 Three-dimensional structure prediction of fusion protein
根据融合蛋白的核苷酸序列,我们利用http://raptorx.uchicago.edu/网站对XCL1以及VEGFR2胞外区融合蛋白的三维结构进行预测。XCL1属于趋化因子,其趋化功能的正常发挥需要保有完整的空间结构。为了确保XCL1与VEGFR2胞外区融合后,XCL1仍然保留有自身的空间结构,我们对融合后的氨基酸序列在http://raptorx.uchicago.edu/网站上进行三维结构预测,结果如图2显示,XCL1与VEGFR2融合后,二者仍然各自保有原来的空间结构,并不影响XCL1的趋化功能发挥。According to the nucleotide sequence of the fusion protein, we used the website http://raptorx.uchicago.edu/ to predict the three-dimensional structure of the fusion protein of XCL1 and VEGFR2 extracellular domain. XCL1 is a chemokine, and the normal function of its chemotactic function needs to maintain a complete spatial structure. In order to ensure that XCL1 still retains its own spatial structure after the fusion of XCL1 and the extracellular domain of VEGFR2, we predicted the three-dimensional structure of the fused amino acid sequence on the website http://raptorx.uchicago.edu/, and the results are shown in Figure 2. After the fusion of XCL1 and VEGFR2, the two still retain their original spatial structure, which does not affect the chemotactic function of XCL1.
实施例3 编码XCL1-VEGFR2融合蛋白的哺乳动物细胞表达载体表达效果检测Example 3 Detection of expression effect of mammalian cell expression vector encoding XCL1-VEGFR2 fusion protein
转染前24小时,在6孔细胞培养板内接种1*10 6个HEK293T细胞,待细胞密度长到70%-80%时开始转染试验。转染时提前在37°水浴锅中预热细胞培养基、无血清的Opti-MEM培养基。转染时将5微克空载体(Vector),单独VEGFR2表达载体,XCL1-VEGFR2野生型和突变型融合基因质粒和20μL PEI转染试剂先后加入到200μL无血清的Opti-MEM中,混合均匀后,室温下静置10分钟。将需要转染的细胞更换新鲜培养基,轻柔加入上述转染体系,轻轻摇匀。将细胞放回细胞培养 箱中培养6小时后换液。转48小时后收取细胞用Western Blot检测XCL1-VEGFR2融合基因质粒HEK293T细胞中的表达效果。 24 hours before transfection, inoculate 1*10 6 HEK293T cells in a 6-well cell culture plate, and start the transfection experiment when the cell density reaches 70%-80%. Before transfection, preheat the cell culture medium and serum-free Opti-MEM medium in a 37° water bath in advance. During transfection, add 5 micrograms of empty vector (Vector), VEGFR2 expression vector alone, XCL1-VEGFR2 wild-type and mutant fusion gene plasmids, and 20 μL of PEI transfection reagent into 200 μL of serum-free Opti-MEM, and mix them evenly. Let stand at room temperature for 10 minutes. Replace the cells to be transfected with fresh medium, gently add the above transfection system, and shake gently. Put the cells back into the cell incubator and culture for 6 hours, then change the medium. After 48 hours of transfection, the cells were collected and Western Blot was used to detect the expression effect of XCL1-VEGFR2 fusion gene plasmid HEK293T cells.
为了方便检测融合基因的表达效果我们在融合蛋白的C端连接一个DDDDK5个氨基酸组成的Flag标签,以便利用Flag标签抗体检测融合蛋白的表达。收集细胞,加入60μL的含有PMSF或Cocktail蛋白酶抑制剂的0.5%NP40裂解缓冲液。充分重悬细胞,4℃旋转裂解细胞30分钟。12000rpm,4℃离心裂解液10分钟,收集上清至新的1.5mL EP管中,弃掉沉淀。根据样品实际体积加入5×SDS-PAGE蛋白上样缓冲液,混合均匀后将样品放在100℃空气浴中加热10分钟,立即进行Western blot,利用Flag标签抗体(Sigma,F3165)进行检测,结果如图3显示空载体(Vector)无蛋白表达,单独VEGFR2表达载体,XCL1-VEGFR2野生型和突变型融合基因质粒均能有效表达并且表达量相当。In order to facilitate the detection of the expression effect of the fusion gene, we attached a 5-amino acid Flag tag composed of DDDDK to the C-terminus of the fusion protein, so that the Flag tag antibody can be used to detect the expression of the fusion protein. Collect the cells and add 60 μL of 0.5% NP40 lysis buffer containing PMSF or Cocktail protease inhibitor. Fully resuspend the cells and spin at 4°C to lyse the cells for 30 minutes. Centrifuge the lysate at 12000 rpm and 4°C for 10 minutes, collect the supernatant into a new 1.5 mL EP tube, and discard the precipitate. Add 5×SDS-PAGE protein loading buffer according to the actual volume of the sample. After mixing, heat the sample in an air bath at 100°C for 10 minutes, and then perform Western blot immediately. Use Flag tag antibody (Sigma, F3165) for detection. Figure 3 shows that the empty vector (Vector) has no protein expression, and the VEGFR2 expression vector alone, XCL1-VEGFR2 wild-type and mutant fusion gene plasmids can be effectively expressed and the expression amount is equivalent.
实施例4 编码XCL1-VEGFR2融合蛋白的昆虫杆状细胞表达载体表达以及蛋白纯化Example 4 Expression of insect rod cell expression vector encoding XCL1-VEGFR2 fusion protein and protein purification
细胞和培养基的选择:本实验所用的细胞表达是昆虫细胞SF9(草地贪夜蛾细胞),此细胞可以贴壁或悬浮培养,一般悬浮培养SF9细胞进行目的蛋白的大量表达。培养基为无血清培养基,本实验病毒包被阶段需用Gibico培养基贴壁培养。重组蛋白的大量制备阶段选用的是义翘神州SIM SF培养基悬浮培养。Selection of cells and culture medium: The cell expression used in this experiment is insect cell SF9 (Spodoptera frugiperda). This cell can be cultured in attachment or suspension. Generally, SF9 cells are cultured in suspension to express a large amount of the target protein. The medium is a serum-free medium, and the Gibico medium should be used for adherent culture during the virus coating stage of this experiment. For the mass preparation stage of recombinant protein, the suspension culture of Yiqiao Shenzhou SIM SF medium is used.
(1)转染昆虫细胞(1) Transfection of insect cells
准备细胞:使用Polystyrene的6孔培养板,细胞总数为0.9×10 6细胞/孔,多次轻轻上下和左右来回移动使细胞均匀地铺在培养板上,静置30min; Prepare the cells: use a polystyrene 6-well culture plate, the total number of cells is 0.9×10 6 cells/well, move gently up and down and left and right to make the cells evenly spread on the culture plate, and let it stand for 30 minutes;
(2)DNA/lipid混合物的制备:(2) Preparation of DNA/lipid mixture:
1)向1.5ml EP管中加入100μl Gibco培养基,再加入1μl Genejuice,此为溶液1;1) Add 100μl Gibco medium to a 1.5ml EP tube, then add 1μl Genejuice, this is solution 1;
2)向另一个1.5ml EP管中先加入100μl Gibco培养基,再加入1μg Bacmid,此为溶液2;2) Add 100μl Gibco medium to another 1.5ml EP tube, and then add 1μg Bacmid, this is solution 2;
3)使溶液1和溶液2充分混合,室温下孵育30min;3) Mix Solution 1 and Solution 2 thoroughly, and incubate for 30 minutes at room temperature;
(3)转染:再加800μl Gibco培养基到DNA/lipid混合物,把培养板中的细胞培养液吸干,快速把1ml DNA/lipid混合物转至细胞单层上,轻轻摇晃均匀,27℃静置5个小时;(3) Transfection: Add 800μl of Gibco medium to the DNA/lipid mixture, blot the cell culture medium in the culture plate, and quickly transfer 1ml of the DNA/lipid mixture to the cell monolayer. Shake gently, 27°C Let stand for 5 hours;
(4)培养:5个小时之后,去掉转染培养基,再加入2ml Gibco培养基,27℃静置培养;(4) Cultivation: After 5 hours, remove the transfection medium, add 2ml Gibco medium, and cultivate at 27°C;
(5)孵育72小时后能观察到病毒感染的现象,在第5~6天提取P0病毒株。(5) Virus infection can be observed after 72 hours of incubation, and the P0 virus strain is extracted on the 5th to 6th day.
(6)病毒株制备、扩增及鉴定(6) Preparation, amplification and identification of virus strains
1)制备病毒株:一旦得到转染细胞且出现晚期转染现象,则可从6孔板中的孔中收集含有病毒的细胞,并转入无菌的2.0ml离心管中。离心5min除去细胞和碎片。之后将上清转到新的2.0ml离心管中,即P0病毒株,加入10%FBS,4℃避光储存。1) Preparation of virus strains: Once the transfected cells are obtained and late-stage transfection occurs, the virus-containing cells can be collected from the wells in the 6-well plate and transferred to a sterile 2.0 ml centrifuge tube. Centrifuge for 5 min to remove cells and debris. Then transfer the supernatant to a new 2.0ml centrifuge tube, which is the P0 virus strain, add 10% FBS, and store at 4°C in the dark.
2)扩增病毒株:取100~200μl P0病毒加入到总细胞数为18~20×10 6、总体积为15ml的培养基中,用T75进行病毒扩增;培养4~5天后收集含有病毒的培养基,并转入无菌的15ml离心管中。500×g离心5min,分别收集上清和沉淀 2) Amplify the virus strain: add 100~200μl of P0 virus to a medium with a total cell number of 18~20×10 6 and a total volume of 15ml, and use T75 for virus amplification; collect the virus after culturing for 4~5 days And transferred to a sterile 15ml centrifuge tube. Centrifuge at 500×g for 5 min, and collect the supernatant and precipitate separately
(7)重组蛋白的大量制备(7) Mass production of recombinant protein
1)病毒感染:利用昆虫细胞SF9表达目的蛋白,细胞扩增至合适感染时的细胞密度,加入相应量的病毒。大约65h后收集细胞悬液。1) Virus infection: Use insect cells SF9 to express the target protein, expand the cells to the appropriate cell density for infection, and add the corresponding amount of virus. After about 65 hours, the cell suspension was collected.
2)细胞上清液收集:目的蛋白为分泌蛋白,因此目的蛋白在上清液中。将表达好目的蛋白的细胞悬液使用高速离心机8000rpm/4℃离心30min,使细胞与上清液分离,取上清液,之后直接进行体外亲和层析的蛋白纯化。2) Collection of cell supernatant: The target protein is a secreted protein, so the target protein is in the supernatant. The cell suspension expressing the target protein was centrifuged at 8000 rpm/4°C for 30 min in a high-speed centrifuge to separate the cells from the supernatant, and then the supernatant was taken, and then directly subjected to in vitro affinity chromatography for protein purification.
3)利用多聚组氨酸标签(His-tag)进行亲和层析的蛋白纯化构建的目的蛋白的N-端带有由连续六个组氨酸组成的His-tag,组氨酸侧链的咪唑基团能够结合金属离子(镍或钴)。如果将镍或钴离子固定在层析介质上,当含有His-tag融合蛋白的混合液流经层析柱时His-tag融合蛋白被吸附在柱子上,杂蛋白全部流出;之后使用较高浓度的咪唑溶液(如 200mM)洗脱,咪唑便与His-tag的咪唑环竞争,最终将His-tag融合蛋白洗脱下来。组氨酸标签很小且几乎不会影响目的蛋白的功能、活性或结构,应用比较广泛。主要实验步骤如下所示:3) Protein purification constructed by affinity chromatography using polyhistidine tag (His-tag) has a His-tag composed of six consecutive histidines on the N-terminus of the target protein, histidine side chain The imidazole group can bind metal ions (nickel or cobalt). If the nickel or cobalt ions are immobilized on the chromatography medium, when the mixed solution containing the His-tag fusion protein flows through the chromatography column, the His-tag fusion protein is adsorbed on the column, and all the miscellaneous proteins flow out; then use a higher concentration The imidazole solution (such as 200 mM) is eluted, and the imidazole will compete with the imidazole ring of His-tag, and finally the His-tag fusion protein will be eluted. The histidine tag is very small and hardly affects the function, activity or structure of the target protein, so it is widely used. The main experimental steps are as follows:
1)结合:将离心后获得的细胞上清液加入到镍柱中,于4℃结合将上清液缓慢流穿Ni Beads一遍,使带有His标签的目的蛋白与Ni Beads充分结合;1) Binding: add the cell supernatant obtained after centrifugation to the nickel column, combine at 4°C, and slowly flow the supernatant through the Ni Beads to make the target protein with the His tag fully bind to the Ni Beads;
2)洗涤杂蛋白:先用加入了20mM咪唑的PBS溶液进行洗涤,洗涤过程中使用考马斯亮蓝染液中检测杂蛋白是否洗涤完全;2) Wash the contaminated protein: first wash with a PBS solution added with 20mM imidazole, and use Coomassie brilliant blue staining solution to check whether the contaminated protein is completely washed during the washing process;
3)洗脱目的蛋白:用加入了200mM咪唑的PBS洗脱液洗脱目的蛋白,洗脱过程中使用考马斯亮蓝染液中检测是否洗涤完全;3) Elution of the target protein: The target protein is eluted with the PBS elution solution added with 200 mM imidazole, and the Coomassie brilliant blue staining solution is used to check whether the washing is complete during the elution process;
4)把步骤3洗脱得到的目的蛋白溶液用4℃离心机进行浓缩至终体积500μl。4) Concentrate the target protein solution eluted in step 3 with a 4°C centrifuge to a final volume of 500 μl.
5)凝胶过滤层析柱的分离纯化:把步骤4得到的浓缩液上样至已经以PBS预先平衡好的凝胶过滤层析柱Superdex 200,收集目的蛋白峰,用Western Blot鉴定是否为目的蛋白,并且利用12%SDS-PAGE电泳鉴定蛋白纯度,如图4所示;5) Separation and purification of gel filtration chromatography column: apply the concentrated solution obtained in step 4 to the gel filtration chromatography column Superdex 200 that has been pre-balanced with PBS, collect the target protein peak, and use Western Blot to identify whether it is the purpose Protein, and the purity of the protein was identified by 12% SDS-PAGE electrophoresis, as shown in Figure 4;
6)浓缩冻存:根据12%SDS-PAGE电泳鉴定的结果,将纯度达到要求的蛋白进行浓缩,利用微量分光光度计测定蛋白浓度后分装,液氮速冻保存于-80℃。6) Concentration and cryopreservation: According to the results of 12% SDS-PAGE electrophoresis identification, the protein with the required purity is concentrated, and the protein concentration is measured by a micro-spectrophotometer, and then aliquoted, and stored in liquid nitrogen quick freezing at -80°C.
实施例5,XCL1-VEGFR2融合蛋白结合MHC-II+CD11c+CD8a+抗原交叉呈递DC细胞能力检测Example 5: Detection of the ability of XCL1-VEGFR2 fusion protein to bind MHC-II+CD11c+CD8a+ antigen to cross-present DC cells
获得纯化后的融合蛋白后,本发明首先对从昆虫细胞中获得的蛋白是否可以有效地结合MHCII+CD11c+CD8a+抗原交叉呈递DC细胞进行了验证。After obtaining the purified fusion protein, the present invention first verifies whether the protein obtained from insect cells can effectively bind to MHCII+CD11c+CD8a+ antigen cross-presenting DC cells.
(1)分离纯化CD11c+DC细胞(1) Isolation and purification of CD11c+DC cells
以小鼠全脾作为CD11c+DC细胞来源,先取小鼠脾利用70um孔径细胞筛网在1640培养基中磨碎成单细胞,200g离心10分钟后,弃上清,利用R&D公司红细胞裂解液(货号WL2000)对脾细胞进行裂红处理。 具体为先将裂解液A用蒸馏水稀释10倍配成工作液,每个脾加入2mL工作液重悬,室温放置10min,期间将中和液B用蒸馏水稀释10倍配成工作液。十分钟后将10mL中和液B加入裂解液中中和后200g离心10min。细胞团用含1%灭活FBS的PBS缓冲液洗涤一次并计数。取总数1*10 8次方个细胞重悬于400μL含1%灭活FBS的PBS缓冲液中,加入100μLCD11c磁珠(Miltenyi Biotec:130-108-338),4°避光放置20min。这期间利用1%灭活FBS的PBS缓冲液平衡吸附磁珠的柱子。20min后将细胞以及磁珠混合液一并转入磁珠吸附柱子,并将柱子放于磁力架上,待细胞完全进入柱子中,用1%灭活FBS的PBS缓冲液洗涤三次每次3mL。之后将柱子从磁力架取下放于15mL离心管上方加入5mL1%灭活FBS的PBS缓冲液并快速推动将CD11c阳性DC细胞洗脱,计数并用200g离心10min。将最终获得的CD11c阳性DC细胞用无血清1640重悬为1*10 6每mL。铺于24孔板中,每孔1mL。将细胞分为BSA蛋白对照组,VEGFR2蛋白对照组以及XCL1-VEGFR2野生型蛋白实验组,每组加入蛋白总量为50μg混匀后放于37°二氧化碳培养箱培养40分钟后,收集细胞500g离心,并用1%灭活FBS的PBS缓冲液反复洗涤两次。并进行流式染色:MHC-II APC,Flag-dye light 488,CD8α-Percp/Cy5.5。利用BD LSRII仪器中速收集细胞,并分析不同组之间MHC-II+Flag-dye light 488+CD8α+细胞比例。每组重复3次。 Using mouse whole spleen as the source of CD11c+DC cells, first take mouse spleen and use 70um pore size cell mesh to grind into single cells in 1640 medium. After centrifugation at 200g for 10 minutes, discard the supernatant and use R&D company red blood cell lysate ( Catalog No. WL2000) spleen cells were treated with split red. Specifically, the lysis solution A was diluted 10 times with distilled water to prepare a working solution, and each spleen was resuspended by adding 2 mL of working solution, and placed at room temperature for 10 minutes, during which the neutralization solution B was diluted with distilled water 10 times to prepare a working solution. Ten minutes later, add 10 mL of neutralization solution B to the lysis solution and centrifuge at 200 g for 10 minutes after neutralization. The cell pellet was washed once with PBS buffer containing 1% inactivated FBS and counted. Take a total of 1*10 8 cells and resuspend them in 400 μL of PBS buffer containing 1% inactivated FBS, add 100 μ LCD11c magnetic beads (Miltenyi Biotec: 130-108-338), and place them in the dark at 4° for 20 minutes. During this period, 1% FBS-inactivated PBS buffer was used to equilibrate the column that adsorbed the magnetic beads. After 20 minutes, the cells and the magnetic bead mixture were transferred to the magnetic bead adsorption column together, and the column was placed on the magnetic stand. After the cells were completely inserted into the column, the cells were washed three times with 3 mL of 1% inactivated FBS PBS buffer. Afterwards, the column was removed from the magnetic stand and placed on the top of the 15 mL centrifuge tube. 5 mL of 1% FBS-inactivated PBS buffer was added and quickly pushed to elute the CD11c-positive DC cells, counted and centrifuged at 200 g for 10 min. The finally obtained CD11c-positive DC cells were resuspended to 1*10 6 per mL with serum-free 1640. Spread in a 24-well plate, 1mL per well. The cells were divided into BSA protein control group, VEGFR2 protein control group and XCL1-VEGFR2 wild-type protein experimental group. The total amount of protein in each group was 50μg, mixed well, and incubated in a 37° carbon dioxide incubator for 40 minutes. The cells were collected and centrifuged at 500g. , And washed twice with 1% inactivated FBS PBS buffer. And perform flow dyeing: MHC-II APC, Flag-dye light 488, CD8α-Percp/Cy5.5. Use BD LSRII instrument to collect cells at medium speed, and analyze the ratio of MHC-II+Flag-dye light 488+CD8α+ cells between different groups. Repeat 3 times for each group.
(2)结果显示,未融合XCL1的VEGFR2蛋白与BSA对照蛋白结合的MHC-II+CD11c+CD8a+数目以及比例类似,而融合了XCL1的VEGFR2蛋白更强的结合到MHC-II+CD11c+CD8a+抗原交叉呈递DC细胞上,结合能力高于对照组20倍以上,如图5A,5B所示。(2) The results show that the number and proportion of MHC-II+CD11c+CD8a+ bound to the VEGFR2 protein without XCL1 and the BSA control protein are similar, while the VEGFR2 protein fused with XCL1 binds to the MHC-II+CD11c+CD8a+ antigen more strongly On cross-presenting DC cells, the binding capacity is more than 20 times higher than that of the control group, as shown in Figures 5A and 5B.
实施例6 XCL1-VEGFR2融合蛋白对MHCII+CD11c+CD8a+抗原交叉呈递DC细胞的趋化能力检测Example 6 Detection of the chemotaxis ability of XCL1-VEGFR2 fusion protein on MHCII+CD11c+CD8a+ antigen cross-presenting DC cells
(1)鉴于XCL1-VEGFR2融合蛋白可以有效地结合MHCII+CD11c+CD8a+抗原交叉呈递DC细胞,我们根据以往研究中表述XCL1趋化因子可以有效招募MHCII+CD11c+CD8a+抗原交叉呈递DC细 胞,设计Tanswell试验验证融合蛋白可以有效地趋化招募DC细胞。Transwell试验采用上下两层的双层细胞培养板,下层加入趋化因子,上层铺入靶细胞,在本实验中为经CD11c磁珠富集过的DC细胞。上层培养室底部为特定孔径的膜,本实验根据既往研究报道选用24孔Transwell板上层直径6.5mm,5μm孔径的膜,将靶细胞铺于膜上,将会由于下层趋化因子的吸引而穿透膜进入下层。进入下层细胞的多少与趋化因子的趋化能力成正比。我们首先将经CD11c磁珠富集过的DC细胞重悬为1*10 7每mL,在上层小室中加入100μL细胞即1*10 6个。在下层加入含有BSA蛋白70ng的对照组,VEGFR2蛋白70ng对照组以及XCL1-VEGFR2野生型和突变体蛋白70ng的实验组培养基700μL,以保证上下层培养基的液面高度一致,防止上层细胞因为液面高度不同而因压强作用进入到下层中。将培养板放于二氧化碳培养箱培养5小时。去掉上层小室收集下层培养基中细胞,500g离心,并用1%灭活FBS的PBS缓冲液反复洗涤两次。并进行流式染色:MHC-II APC,CD11c-PE,CD8α-Percp/Cy5.5。利用BD Canto II仪器中速收集细胞,并分析不同组之间MHC-II+CD11c-PE+CD8α+细胞比例。每组重复3次。 (1) In view of the fact that XCL1-VEGFR2 fusion protein can effectively bind to MHCII+CD11c+CD8a+ antigen cross-presenting DC cells, based on previous studies that XCL1 chemokine can effectively recruit MHCII+CD11c+CD8a+ antigen cross-presenting DC cells, we designed Tanswell Experiments verify that the fusion protein can effectively chemotactic and recruit DC cells. The Transwell test uses a double-layered cell culture plate with upper and lower layers, the lower layer is added with chemokines, and the upper layer is covered with target cells. In this experiment, it is DC cells enriched with CD11c magnetic beads. The bottom of the upper culture chamber is a membrane with a specific pore size. In this experiment, a 24-well Transwell membrane with a diameter of 6.5 mm and a pore size of 5 μm was selected according to previous research reports. Transmembrane enters the lower layer. The number of cells entering the lower layer is directly proportional to the chemotactic ability of the chemokine. We first resuspend the DC cells enriched with CD11c magnetic beads to 1* 107 per mL, and add 100μL of cells to the upper chamber, namely 1*10 6 cells. In the lower layer, 700μL of the control medium containing 70ng of BSA protein, 70ng of VEGFR2 protein, and 70ng of XCL1-VEGFR2 wild-type and mutant proteins of the experimental group were added to ensure that the liquid level of the upper and lower media was consistent and prevent the upper cells from The liquid level is different and enters the lower layer due to pressure. Place the culture plate in a carbon dioxide incubator for 5 hours. Remove the upper chamber to collect the cells in the lower culture medium, centrifuge at 500 g, and wash twice with 1% FBS-inactivated PBS buffer. And perform flow staining: MHC-II APC, CD11c-PE, CD8α-Percp/Cy5.5. Use BD Canto II instrument to collect cells at medium speed, and analyze the ratio of MHC-II+CD11c-PE+CD8α+ cells between different groups. Repeat 3 times for each group.
(2)结果显示,未融合XCL1的VEGFR2蛋白与BSA对照蛋白趋化下来的MHC-II+CD11c+CD8a+数目以及比例类似,而融合了XCL1的VEGFR2蛋白无论是野生型还是突变体均能趋化更多的的MHC-II+CD11c+CD8a+抗原交叉呈递DC细胞,经统计检验具有显著性差异,统计方法为Unpaired t test,如图6A,6B所示。(2) The results show that the number and ratio of MHC-II+CD11c+CD8a+ chemotaxis of VEGFR2 protein without XCL1 and BSA control protein are similar, while VEGFR2 protein fused with XCL1 can chemoattract both wild-type and mutants. More MHC-II+CD11c+CD8a+ antigen cross-presenting DC cells have statistically significant differences. The statistical method is Unpaired test, as shown in Figures 6A and 6B.
实施例7 融合基因对B16黑色素瘤细胞同种移植瘤发生的干预效果Example 7 Intervention effect of fusion gene on the occurrence of B16 melanoma cell allograft tumor
鉴于融合基因在哺乳动物细胞可以正常表达,融合蛋白也可以有效地趋化、结合MHC-II+CD11c+CD8a+抗原交叉呈递DC细胞。我们提取单独VEGFR2和XCL1-VEGFR2野生型和突变型质粒,利用Wealtec公司的基因枪(GDS-80)对小鼠进行免疫质粒注射。并同种移植B16黑色素瘤细胞后观察融合基因免疫对B16黑色素瘤细胞生长的抑制情况。In view of the normal expression of the fusion gene in mammalian cells, the fusion protein can also effectively chemoattract and combine with MHC-II+CD11c+CD8a+ antigen to cross-present DC cells. We extracted separate VEGFR2 and XCL1-VEGFR2 wild-type and mutant plasmids, and used Wealtec's gene gun (GDS-80) to inject immune plasmids into mice. And after allotransplantation of B16 melanoma cells, the inhibition of the growth of B16 melanoma cells by fusion gene immunization was observed.
(1)B16黑色素瘤细胞培养以及成瘤后VEGFR2基因表达情况检测 复苏B16细胞:预热细胞培养基(细胞培养基由Corning公司的DMEM培养基加入10%浓度的FBS配制而成)。从液氮或-80℃冰箱中取出待复苏的冻存细胞,快速置37℃水浴锅中融化。细胞溶解完全后,1500rpm离心3min,同时在培养皿中加入8mL预热后的细胞培养基。离心后弃上清,加入1mL预热的细胞培养基,重悬细胞并转移至准备好的培养皿中,标记细胞名称、日期及传代次数。将细胞置于37℃,5%CO2培养箱中培养。第二天传代,预热细胞培养基、胰蛋白酶、PBS缓冲液。用真空泵将培养皿中的旧培养基吸出。加入7mL PBS缓冲液润洗一遍细胞,吸出PBS。加入1mL胰蛋白酶,在37℃孵箱中放置消化恰当时间。加入3mL培养基终止消化,用电动移液器吹打细胞至单细胞悬液并转移至15mL离心管,1500rpm离心3min,同时准备新的培养皿,各自加入8mL培养基。离心后弃上清,根据所需传代比例加入适量培养基重悬细胞,吹打至单细胞悬液,取1mL单细胞悬液加入新的培养皿中,放回5%CO2的37℃培养箱中继续培养。待细胞长到80%-90%密度,按上述办法消化细胞计数并重悬于无血清DMEM培养基中,根据前期我们的成瘤经验,将细胞浓度调整为1*10 6个每mL,每只小鼠腋下皮下接种100μL细胞。两周后取所形成的黑色素瘤和周边正常皮肤,提取总RNA。收取适量的小鼠组织,加入1mL Trizol(Invitrogen)。用组织匀浆仪裂解2mins,4℃转动30mins进行彻底裂解。每1mL Trizol中加入200μL氯仿,剧烈震荡15s,室温静置15mins。预冷4℃离心机,12000rpm,离心10mins。离心后,将上层水相转移到新的1.5mL EP管中,加入500μL异丙醇,颠倒混匀后离心,12000rpm,4℃,离心10mins。离心后可见白色沉淀,弃上清,用DEPC水配置的75%乙醇洗涤RNA沉淀后7500rpm,室温离心5mins。弃上清,室温放置干燥RNA至沉淀呈半透明状态。取适量体积的DEPC水溶解RNA。混匀溶解后的RNA,使用Nanodrop检测RNA的浓度和纯度,在EP管壁及盖子上标记RNA种属类型、浓度、提取时间。利用下表所示引物(SEQ ID No.21~24)进行real time PCR检测。结果显示与周边正常皮肤相比,所形成的黑色素瘤中VEGFR2表达水平显著升高,如图7所示,说 明利用B16细胞评价融合基因疫苗是合理的。 (1) B16 melanoma cell culture and detection of VEGFR2 gene expression after tumorigenesis Resuscitate B16 cells: pre-warm cell culture medium (cell culture medium is prepared by adding 10% FBS to Corning's DMEM medium). Take out the frozen cells to be resuscitated from liquid nitrogen or -80°C refrigerator, and quickly melt them in a 37°C water bath. After the cells were completely lysed, centrifuged at 1500 rpm for 3 min, and at the same time added 8 mL of pre-warmed cell culture medium to the petri dish. After centrifugation, the supernatant was discarded, and 1 mL of pre-warmed cell culture medium was added. The cells were resuspended and transferred to a prepared petri dish. The cell name, date, and number of passages were marked. The cells were cultured in a 37°C, 5% CO2 incubator. Passage the next day, pre-warm the cell culture medium, trypsin, and PBS buffer. Use a vacuum pump to suck out the old medium in the petri dish. Add 7mL PBS buffer to rinse the cells once and aspirate the PBS. Add 1mL trypsin and place it in a 37°C incubator for proper time for digestion. Add 3mL culture medium to terminate the digestion, pipette the cells to a single cell suspension with an electric pipette and transfer to a 15mL centrifuge tube, centrifuge at 1500rpm for 3min, and prepare a new culture dish at the same time, add 8mL culture medium to each. After centrifugation, discard the supernatant, add appropriate amount of medium according to the required passage ratio to resuspend the cells, pipette to a single cell suspension, take 1 mL of the single cell suspension into a new culture dish, and put it back into the 37°C incubator with 5% CO2 Continue to cultivate. When the cells grow to 80%-90% density, digest the cell count according to the above method and resuspend it in serum-free DMEM medium. According to our previous experience of tumor formation, adjust the cell concentration to 1*10 6 cells per mL, each 100 μL of cells were subcutaneously inoculated into the armpits of the mice. Two weeks later, the formed melanoma and surrounding normal skin were taken, and total RNA was extracted. Collect an appropriate amount of mouse tissue and add 1 mL Trizol (Invitrogen). Lysis with a tissue homogenizer for 2mins, and rotate for 30mins at 4°C for complete lysis. Add 200μL of chloroform to each 1mL of Trizol, shake vigorously for 15s, and let stand at room temperature for 15mins. Pre-cooled centrifuge at 4℃, 12000rpm, centrifugation for 10mins. After centrifugation, transfer the upper aqueous phase to a new 1.5mL EP tube, add 500μL of isopropanol, mix upside down and centrifuge at 12000rpm, 4°C for 10mins. A white precipitate can be seen after centrifugation, the supernatant is discarded, and the RNA precipitate is washed with 75% ethanol in DEPC water, and centrifuged at 7500 rpm for 5 mins at room temperature. Discard the supernatant, and place the dried RNA at room temperature until the precipitate is translucent. Take an appropriate volume of DEPC water to dissolve RNA. Mix the dissolved RNA, use Nanodrop to detect the concentration and purity of the RNA, and mark the RNA species type, concentration, and extraction time on the EP tube wall and lid. The primers (SEQ ID No. 21-24) shown in the table below were used for real-time PCR detection. The results showed that compared with surrounding normal skin, the expression level of VEGFR2 in the formed melanoma was significantly increased. As shown in Figure 7, it is reasonable to use B16 cells to evaluate the fusion gene vaccine.
表2Table 2
Figure PCTCN2020109970-appb-000002
Figure PCTCN2020109970-appb-000002
(2)融合基因免疫后对B16黑色素瘤细胞同种移植瘤发生的预防效果(2) The preventive effect of fusion gene immunization on the occurrence of B16 melanoma cell allograft tumors
确定B16细胞成瘤模型可用后,我们按照图8A的时间轴标注的免疫策略对小鼠进行基因枪质粒注射。将C57B6(购自为通利华)周龄雄性小鼠,分为注射单独VEGFR2和XCL1-VEGFR2野生型和突变型质粒的三组,每组五只,利用脱毛膏进行小鼠右侧靠近腹股沟淋巴结处脱毛处理。之后利用基因枪在脱毛处注射质粒,每只50μg,每周一次,共注射四次,在最后一次注射后一周接种之前摸索好成瘤条件的B16黑色素瘤细胞,观察肿瘤形成时间并每两天测量肿瘤的长径a和短径b,按照a*b*b/2进行肿瘤体积计算,绘制肿瘤生长曲线。结果如图8A,8B所示,免疫注射XCL1-VEGFR2野生型和突变型质粒均能有效地延迟肿瘤的发生时间以及生长速度。说明融合基因免疫的治疗效果显著有效。After confirming that the B16 cell tumorigenesis model is available, we injected the mice with gene gun plasmids according to the immunization strategy marked on the time axis in Figure 8A. C57B6 (purchased from Tonglihua) week-old male mice were divided into three groups injected with separate VEGFR2 and XCL1-VEGFR2 wild-type and mutant plasmids, five in each group, and depilatory cream was used to carry out the right side of the mouse near the groin Hair removal treatment at the lymph nodes. After that, use a gene gun to inject the plasmid into the hair removal site, each 50μg, once a week, a total of four injections, one week after the last injection, the B16 melanoma cells with good tumorigenic conditions were explored, and the tumor formation time was observed every two days. Measure the long axis a and short axis b of the tumor, calculate the tumor volume according to a*b*b/2, and draw the tumor growth curve. The results are shown in Figures 8A and 8B. Both the wild-type and mutant-type plasmids of XCL1-VEGFR2 can effectively delay the onset and growth rate of tumors. It shows that the therapeutic effect of fusion gene immunization is significantly effective.
(3)探索融合基因免疫是否诱导出了更强的细胞特异性T细胞反应在对照组肿瘤生长至伦理终点2000mm 3时,处死小鼠将各组小鼠所负荷的肿瘤取下拍照,之后用100μm孔径筛网将细胞研磨成单细胞悬液,800g离心后将细胞重悬与80%percoll溶液中每1000mm 3加4mL重悬,在上层小心加入40%pecoll溶液,800g离心20分钟,弃去最上层肿瘤细胞留中间层淋巴细胞,洗涤计数,最后将细胞重悬于含10%热灭活FBS的1640培养基中浓度为1*10 6个每mL,将细胞铺于24孔板中,每孔1mL并加入VEGFR2表位多肽刺激,同时加入Golgi stop(Biolegend)抑制胞内因子分泌。12小时后收取细胞用1%灭活FBS的PBS缓冲液洗涤细胞两次, 并进行流式染色:Gran B-PE,CD8α-Percp/Cy5.5。结果如图9所示,经XCL1-VEGFR2野生型和突变型质粒免疫后的小鼠肿瘤内针对VEGFR2的特异性T细胞与单独VEGFR2免疫组相比显著增多,说明XCL1-VEGFR2野生型和突变型质粒免疫确实诱导出了特异性反应的CD8a细胞毒性T淋巴细胞。 (3) Explore whether the fusion gene immunization induces a stronger cell-specific T cell response. When the tumor in the control group grew to the ethical endpoint of 2000 mm 3 , the mice were sacrificed and the tumors loaded by the mice in each group were taken off and photographed. Grind the cells into a single cell suspension with a 100μm mesh screen. After 800g centrifugation, resuspend the cells with 80% percoll solution and add 4mL per 1000mm 3 to resuspend. Carefully add 40% pecoll solution to the upper layer. Centrifuge at 800g for 20 minutes and discard. The uppermost tumor cells are left with the middle layer of lymphocytes, washed and counted, and finally the cells are resuspended in 1640 medium containing 10% heat-inactivated FBS at a concentration of 1*10 6 cells per mL, and the cells are plated in a 24-well plate. 1 mL per well was stimulated by adding VEGFR2 epitope polypeptide, and Golgi stop (Biolegend) was added to inhibit the secretion of intracellular factors. After 12 hours, the collected cells were washed twice with 1% FBS-inactivated PBS buffer, and subjected to flow cytometry: Gran B-PE, CD8α-Percp/Cy5.5. The results are shown in Figure 9. The mouse tumors immunized with XCL1-VEGFR2 wild-type and mutant plasmids have significantly more VEGFR2-specific T cells compared with the VEGFR2 immunization group alone, indicating that XCL1-VEGFR2 wild-type and mutant types Plasmid immunization did induce specific CD8a cytotoxic T lymphocytes.
实施例8:融合蛋白免疫后T细胞过继治疗对黑色素瘤的治疗效果Example 8: Therapeutic effect of T cell adoptive therapy on melanoma after immunization with fusion protein
鉴于融合基因免疫后成功的在小鼠体内诱导出了针对VEGFR2的特异性细胞毒性T淋巴细胞,所以本发明利用从昆虫杆状细胞中纯化的融合蛋白,免疫T细胞供体鼠,在供体鼠内诱导针对VEGFR2的特异性细胞毒性T淋巴细胞,并将其过继到接有B16肿瘤细胞的受体鼠内,看是否可以有效抑制受体鼠的肿瘤的发生发展。In view of the successful induction of specific cytotoxic T lymphocytes against VEGFR2 in mice after immunization with the fusion gene, the present invention uses the fusion protein purified from insect rod cells to immunize the T cell donor mouse. Induce specific cytotoxic T lymphocytes against VEGFR2 in mice, and adopt them to recipient mice with B16 tumor cells to see if it can effectively inhibit the occurrence and development of tumors in recipient mice.
(1)过继T细胞供体鼠的融合蛋白免疫(1) Fusion protein immunization of adoptive T cell donor mice
为了使昆虫杆状细胞纯化的融合蛋白能有效的诱导出针对VEGFR2的特异性细胞毒性T淋巴细胞,本发明团队前期结合既往研究XCL1靶向DC细胞的特点对给药途径进行了探索优化,首先采用静脉给药结果如图10A所示,静脉注射KDR蛋白加LPS佐剂并不能产生抗肿瘤效果。最终确定融合蛋白40微克剂量加Poly:IC 30微克加不完全弗氏佐剂乳化后经皮下免疫,每周一次,共免疫两次可以有效地诱导针对VEGFR2的特异性细胞毒性T淋巴细胞产生,经过继治疗对B16黑色素瘤细胞同种移植瘤产生抑制效果。In order to enable the fusion protein purified from insect rod cells to effectively induce specific cytotoxic T lymphocytes against VEGFR2, the team of the present invention explored and optimized the route of administration based on the characteristics of XCL1 targeting DC cells in the previous study. First, The results of intravenous administration are shown in Figure 10A. Intravenous injection of KDR protein plus LPS adjuvant does not produce anti-tumor effects. Finally, it was determined that the fusion protein 40 micrograms dose plus Poly: IC 30 micrograms plus incomplete Freund’s adjuvant emulsified and subcutaneously immunized, once a week, twice co-immunization can effectively induce the production of specific cytotoxic T lymphocytes against VEGFR2. After subsequent treatment, the B16 melanoma cell allograft has an inhibitory effect.
(2)T细胞过继治疗对黑色素瘤的治疗效果(2) The therapeutic effect of adoptive T cell therapy on melanoma
在第二次经皮下蛋白免疫后一周处死小鼠,取小鼠脾脏按照实施例5中的方法进行研磨,裂红并用CD8a磁珠对细胞毒性T淋巴细胞富集,最后将获得的细胞重悬在无血清1640培养基中,浓度为2*10 7个每mL,对提前两天经尾静脉接种3*10 5个B16黑色素瘤细胞的小鼠再次尾静脉过继获得的T细胞每只200μL共2*10 6个细胞。两周后解剖小鼠取小鼠肺组织于10%多聚甲醛中固定,第二天进行拍照,结果如图10B所示,过继经XCL1-VEGFR2野生型和突变型蛋白免疫后的小鼠的细胞毒性T淋巴细 胞后,受体小鼠的肺部结节显著减少。说明经XCL1-VEGFR2野生型和突变型蛋白免疫后的小鼠的产生了针对VEGFR2的细胞毒性T淋巴细胞,并可在体内有效杀灭表达VEGFR2的黑色素瘤细胞。 The mice were sacrificed one week after the second subcutaneous protein immunization, and the mouse spleens were taken and ground according to the method in Example 5, split red and enriched with CD8a magnetic beads for cytotoxic T lymphocytes, and finally the obtained cells were resuspended In the serum-free 1640 medium, the concentration is 2* 107 cells per mL. For mice that were inoculated with 3*10 5 B16 melanoma cells via the tail vein two days in advance, the T cells obtained by adopting the tail vein again each had a total of 200 μL. 2*10 6 cells. Two weeks later, the mice were dissected and the lung tissues of the mice were fixed in 10% paraformaldehyde. The photos were taken the next day. The results are shown in Figure 10B. After cytotoxic T lymphocytes, the lung nodules of the recipient mice were significantly reduced. It shows that mice immunized with XCL1-VEGFR2 wild-type and mutant proteins produce cytotoxic T lymphocytes against VEGFR2, and can effectively kill melanoma cells expressing VEGFR2 in vivo.
以上对本发明所提供的靶向VEGFR2的转移性癌疫苗进行了详细介绍。本文应用了具体个例对本发明的原理及实施方式进行了阐述,以上实施例的说明只是用于帮助理解本发明的方法及其核心思想。应当指出,对于本技术领域技术人员来说,在不脱离本发明原理的前提下,还可以对本发明进行若干改进和修饰,这些改进和修饰也落入本发明权利要求的保护范围内。The metastatic cancer vaccine targeting VEGFR2 provided by the present invention has been introduced in detail above. This article uses specific examples to illustrate the principle and implementation of the present invention. The description of the above examples is only used to help understand the method and core idea of the present invention. It should be pointed out that for those skilled in the art, without departing from the principle of the present invention, several improvements and modifications can be made to the present invention, and these improvements and modifications also fall within the protection scope of the claims of the present invention.

Claims (10)

  1. 融合蛋白,其特征在于,包括促进肿瘤新生血管生成的VEFGR2(KDR)的能够诱发特异性CD8+T反应的胞外区或其MHCI类分子结合表位优化形式胞外区;接头;以及特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1蛋白;The fusion protein is characterized in that it includes the extracellular region of VEFGR2 (KDR) that promotes tumor angiogenesis and can induce a specific CD8+T response or its MHC class I molecule binding epitope-optimized extracellular region; linker; and specificity Human or mouse XCL1 protein that binds to DC cells with antigen cross-presentation ability;
    其中:in:
    所述促进肿瘤新生血管生成的VEFGR2的能够诱发特异性CD8+T反应的胞外区或其MHCI类分子结合表位优化形式胞外区的氨基酸序列如SEQ ID No.1或2中第20~764位氨基酸所示;所述特异性结合具有抗原交叉呈递能力的DC细胞的人或鼠XCL1蛋白的氨基酸序列如SEQ ID No.3或4的1~114位氨基酸所示。The amino acid sequence of the extracellular region of VEFGR2 that can induce a specific CD8+T response or its MHC class I molecule binding epitope optimized form of the extracellular region of VEFGR2 that promotes tumor angiogenesis is as shown in SEQ ID No. 1 or 2 from 20 to 2 The amino acid at position 764 is shown; the amino acid sequence of the human or murine XCL1 protein that specifically binds to DC cells with antigen cross-presenting ability is shown in amino acids 1 to 114 of SEQ ID No. 3 or 4.
  2. 如权利要求1所述的融合蛋白,其特征在于,所述融合蛋白包括如SEQ ID No.7和8中的1~889位氨基酸所示序列。The fusion protein according to claim 1, wherein the fusion protein comprises a sequence as shown in amino acids 1 to 889 in SEQ ID Nos. 7 and 8.
  3. 编码如权利要求1或2所述的融合蛋白的核苷酸,其特征在于,所述核苷酸的序列:The nucleotide encoding the fusion protein of claim 1 or 2, wherein the sequence of the nucleotide:
    (Ⅰ)、如SEQ ID No.11~16任意所示;(Ⅰ), as shown in SEQ ID No. 11-16;
    or
    (Ⅱ)、如SEQ ID No.11~16任意所示的核苷酸序列的互补核苷酸序列所示;或(II), as shown in the complementary nucleotide sequence of the nucleotide sequence arbitrarily shown in SEQ ID No. 11-16; or
    (Ⅲ)、如与(Ⅰ)或(Ⅱ)的核苷酸序列编码相同蛋白质,但因遗传密码的简并性而与(Ⅰ)或(Ⅱ)的核苷酸序列不同的核苷酸序列所示。(Ⅲ). For example, a nucleotide sequence that encodes the same protein as the nucleotide sequence of (I) or (II), but is different from the nucleotide sequence of (I) or (II) due to the degeneracy of the genetic code Shown.
  4. 重组表达载体,其特征在于,包括载体以及如权利要求1或2所述的融合蛋白或如权利要求3所述的核苷酸。The recombinant expression vector is characterized by comprising a vector and the fusion protein according to claim 1 or 2 or the nucleotide according to claim 3.
  5. 如权利要求4所述的重组表达载体,其特征在于,所述载体包括pcDNA3.1(+)、pcDNA3.1(-)、pFastbac1-dual-MBP。The recombinant expression vector of claim 4, wherein the vector comprises pcDNA3.1(+), pcDNA3.1(-), pFastbac1-dual-MBP.
  6. 包括如权利要求1或2所述的融合蛋白或如权利要求3所述的核苷酸的重组菌株或细胞。A recombinant strain or cell comprising the fusion protein according to claim 1 or 2 or the nucleotide according to claim 3.
  7. 如权利要求1或2所述的融合蛋白、如权利要求3所述的核苷酸、 如权利要求4或5所述的重组表达载体或如权利要求6所述的重组菌株或细胞在制备转移性癌症或VEGFR2高表达的黑色素瘤的疫苗或制备预防和/或治疗转移性癌症或VEGFR2高表达的黑色素瘤的药物中的应用。The fusion protein as claimed in claim 1 or 2, the nucleotide as claimed in claim 3, the recombinant expression vector as claimed in claim 4 or 5, or the recombinant strain or cell as claimed in claim 6 in preparation and transfer The application of a vaccine for a melanoma with high expression of VEGFR2 or a vaccine or a medicine for the prevention and/or treatment of a metastatic cancer or a melanoma with a high expression of VEGFR2.
  8. 如权利要求7所述的应用,其特征在于,所述转移性癌症包括肝癌,结直肠癌或肺癌。The use according to claim 7, wherein the metastatic cancer comprises liver cancer, colorectal cancer or lung cancer.
  9. 转移性癌症或VEGFR2高表达的黑色素瘤的疫苗,其特征在于,包括如权利要求1或2所述的融合蛋白、如权利要求3所述的核苷酸、如权利要求4或5所述的重组表达载体以及药学上可接受的运载体、赋形剂和/或佐剂。A vaccine for metastatic cancer or melanoma with high expression of VEGFR2, characterized by comprising the fusion protein according to claim 1 or 2, the nucleotide according to claim 3, and the vaccine according to claim 4 or 5. Recombinant expression vectors and pharmaceutically acceptable carriers, excipients and/or adjuvants.
  10. 预防和/或治疗转移性癌症或VEGFR2高表达的黑色素瘤的药物,其特征在于,包括如权利要求1或2所述的融合蛋白、如权利要求3所述的核苷酸、如权利要求4或5所述的重组表达载体以及药学上可接受的辅料。A drug for preventing and/or treating metastatic cancer or melanoma with high expression of VEGFR2, characterized by comprising the fusion protein according to claim 1 or 2, the nucleotide according to claim 3, and the fusion protein according to claim 4 Or the recombinant expression vector described in 5 and pharmaceutically acceptable excipients.
PCT/CN2020/109970 2020-04-09 2020-08-19 Vegfr2-targeted metastatic cancer vaccine WO2021203608A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010273819.0 2020-04-09
CN202010273819.0A CN111440244B (en) 2020-04-09 2020-04-09 Metastatic cancer vaccine targeting VEGFR2

Publications (1)

Publication Number Publication Date
WO2021203608A1 true WO2021203608A1 (en) 2021-10-14

Family

ID=71651292

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/109970 WO2021203608A1 (en) 2020-04-09 2020-08-19 Vegfr2-targeted metastatic cancer vaccine

Country Status (2)

Country Link
CN (1) CN111440244B (en)
WO (1) WO2021203608A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111440244B (en) * 2020-04-09 2021-03-16 诺未科技(北京)有限公司 Metastatic cancer vaccine targeting VEGFR2
CN114316025A (en) * 2020-12-25 2022-04-12 百奥赛图(北京)医药科技股份有限公司 VEGFR2 gene humanized non-human animal and construction method and application thereof
CN112979829B (en) * 2021-04-29 2021-08-13 诺未科技(北京)有限公司 Fusion protein and application thereof in preparation of vaccine targeting new coronavirus SARS-COV-2
CN113072648B (en) * 2021-06-04 2021-08-24 诺未科技(北京)有限公司 Liver cancer vaccine targeting AFP
CN113072645A (en) * 2021-06-04 2021-07-06 诺未科技(北京)有限公司 Cancer vaccine targeting EphA2 antigen
CN113072646A (en) * 2021-06-04 2021-07-06 北京承诺未来科技有限公司 EGFR-targeting tumor nucleic acid vaccines

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160074489A1 (en) * 2014-09-15 2016-03-17 Regen Biopharma, Inc Stimulation of immunity to tumor specific and endothelial specific proteins by in vivo dc attraction and maturation
CN107207603A (en) * 2015-02-03 2017-09-26 吉安特科技股份有限公司 Chemotactic factor (CF) immunoglobulin fused polypeptide, its composition, preparation method and purposes
WO2018073185A1 (en) * 2016-10-17 2018-04-26 Vetoquinol Sa Modified antibody constant region
US20180325948A1 (en) * 2015-11-11 2018-11-15 Thomas Ichim Tumor antigen modified dendritic cells
WO2019134018A1 (en) * 2018-01-05 2019-07-11 Telethon Kids Institute Vaccine conjugates and uses thereof
CN111440244A (en) * 2020-04-09 2020-07-24 诺未科技(北京)有限公司 Metastatic cancer vaccine targeting VEGFR2

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SK287357B6 (en) * 1999-02-12 2010-08-09 The Scripps Research Institute Use an alpha v beta 3 antagonist for the preparation of a pharmaceutical composition for treating a tumor cell and therapeutical composition and kit for treating a tumor or tumor metastases
US20060257359A1 (en) * 2005-02-28 2006-11-16 Cedric Francois Modifying macrophage phenotype for treatment of disease
WO2014139468A1 (en) * 2013-03-15 2014-09-18 Admark Healthcare, Llc Fusion protein molecules and method of use
CN109422816B (en) * 2017-08-25 2020-11-20 诺未科技(北京)有限公司 Liver cancer vaccine targeting secondary lymphoid tissue

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160074489A1 (en) * 2014-09-15 2016-03-17 Regen Biopharma, Inc Stimulation of immunity to tumor specific and endothelial specific proteins by in vivo dc attraction and maturation
CN107207603A (en) * 2015-02-03 2017-09-26 吉安特科技股份有限公司 Chemotactic factor (CF) immunoglobulin fused polypeptide, its composition, preparation method and purposes
US20180325948A1 (en) * 2015-11-11 2018-11-15 Thomas Ichim Tumor antigen modified dendritic cells
WO2018073185A1 (en) * 2016-10-17 2018-04-26 Vetoquinol Sa Modified antibody constant region
WO2019134018A1 (en) * 2018-01-05 2019-07-11 Telethon Kids Institute Vaccine conjugates and uses thereof
CN111440244A (en) * 2020-04-09 2020-07-24 诺未科技(北京)有限公司 Metastatic cancer vaccine targeting VEGFR2

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DATABASE Proetein 16 March 2021 (2021-03-16), ANONYMOUS: "lymphotactin precursor [Homo sapiens]", XP055856603, retrieved from GENBANK Database accession no. NP_002986 *
DATABASE Protein 16 October 2021 (2021-10-16), ANONYMOUS: "lymphotactin precursor [Mus musculus]", XP055856612, retrieved from NCBI Database accession no. NP_032536 *
DATABASE Protein 21 October 2008 (2008-10-21), ANONYMOUS: "FLK-1[Mus musculus]", XP055856602, retrieved from GENBANK Database accession no. CAA50192 *
DATABASE Protein 7 October 2020 (2020-10-07), ANONYMOUS: "RecName: Full=Vascular endothelial growth factor receptor 2; Short=VEGFR-2; AltName:Full=Fetal liver kinase 1; Short=FLK-1; AltName: Full=Kinase NYK; AltName: Full=Protein-tyrosine kinase receptor flk-1; AltName: CD_antigen=CD309; Flags: Precursor", XP055856609, retrieved from UniProtKB/Swiss-Prot Database accession no. P35918 *
NATALIA K. BOTELHO , BENJAMIN O. TSCHUMI , JEFFREY A. HUBBELL , MELODY A. SWARTZ , ALENA DONDA , PEDRO ROMERO: "Combination of Synthetic Long Peptides and XCL1 Fusion Proteins Results in Superior Tumor Control", FRONTIERS IN IMMUNOLOGY, vol. 10, 294, 26 February 2019 (2019-02-26), pages 1 - 11, XP055856594, DOI: 10.3389/fimmu.2019.00294 *
WANG SHENGCHAO: "Anti-metastatic Effects of DNA Vaccine Encoding Single-chain Trimer Composed of MHC I And Vascular Endothelial Growth Factor Receptor 2 Peptide", CHINESE MASTER'S THESES FULL-TEXT DATABASE, 1 June 2015 (2015-06-01), pages 1 - 52, XP055856598 *

Also Published As

Publication number Publication date
CN111440244B (en) 2021-03-16
CN111440244A (en) 2020-07-24

Similar Documents

Publication Publication Date Title
WO2021203608A1 (en) Vegfr2-targeted metastatic cancer vaccine
JP4662708B2 (en) Methods and compositions for targeting proteins to exosomes
US7446185B2 (en) Her2/neu target antigen and use of same to stimulate an immune response
PT2155243E (en) Compositions and methods comprising klk3, psca, or folh1 antigen
JP2021536435A (en) Therapeutic agents containing nucleic acids and CAR-modified immune cells and their use
US11648306B2 (en) Non-integrative listeria-based vaccine and method for inducing antitumor immune response
WO2017071173A1 (en) Tumor therapeutic agent modified by il-12/cd62l fusion protein and preparation method and use thereof
CN108866088B (en) Targeting CLL-1 chimeric antigen receptor and uses thereof
CN112574317A (en) Recombinant protein, pharmaceutical composition and application
CN112533939B (en) Tumor immunotherapy composition based on attenuated listeria activated antigen presenting cells, preparation method and application
AU2006327514B2 (en) DNA vaccine for cancer therapy
EP3119422A2 (en) A medicament for use in a method of inducing or extending a cellular cytotoxic immune response
CN108624608B (en) Preparation method and application of fourth generation chimeric antigen receptor targeting mesothelin
CN108624607B (en) Methods and uses of chimeric antigen receptors targeting mesothelin and dual modifications thereof
Bleifuss et al. The translocation motif of hepatitis B virus improves protein vaccination
WO2023137946A1 (en) Use of ccl13
CN108728458B (en) Methods and uses of chimeric antigen receptors targeting mesothelin in combination with IL-15 expression
CA2529058A1 (en) Increased t-cell tumor infiltration by mutant light
CN110845621A (en) Chimeric antigen receptor method targeting EGFR and CD19 double targets
CN110564749B (en) Chimeric antigen receptor targeting EGFR and uses thereof
CN112063640A (en) Chimeric antigen receptor targeting humanized CEA and uses thereof
CN112080510A (en) Chimeric antigen receptor targeting humanized GD2 and uses thereof
CN114790248B (en) MUC1-PDL1-IgG1Fc tumor vaccine and preparation method and application thereof
WO2017107353A1 (en) Cancer treatment agent, preparation method and use thereof employing il-12 with stable membrane expression
WO2021170111A1 (en) Tumor immune enhancer, and preparation method therefor and application thereof

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: 20930612

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20930612

Country of ref document: EP

Kind code of ref document: A1