WO2019128952A1 - 携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用 - Google Patents

携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用 Download PDF

Info

Publication number
WO2019128952A1
WO2019128952A1 PCT/CN2018/123298 CN2018123298W WO2019128952A1 WO 2019128952 A1 WO2019128952 A1 WO 2019128952A1 CN 2018123298 W CN2018123298 W CN 2018123298W WO 2019128952 A1 WO2019128952 A1 WO 2019128952A1
Authority
WO
WIPO (PCT)
Prior art keywords
car
exosomes
cells
cell
immune cell
Prior art date
Application number
PCT/CN2018/123298
Other languages
English (en)
French (fr)
Inventor
胡适
傅文燕
Original Assignee
沣潮医药科技(上海)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 沣潮医药科技(上海)有限公司 filed Critical 沣潮医药科技(上海)有限公司
Priority to US17/043,670 priority Critical patent/US20210030801A1/en
Priority to EP18897524.7A priority patent/EP3747995B1/en
Publication of WO2019128952A1 publication Critical patent/WO2019128952A1/zh

Links

Images

Classifications

    • 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
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464403Receptors for growth factors
    • A61K39/464404Epidermal growth factor receptors [EGFR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464403Receptors for growth factors
    • A61K39/464406Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464424CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • 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
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70517CD8
    • 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
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • 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
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/804Blood cells [leukemia, lymphoma]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/812Breast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/10Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the structure of the chimeric antigen receptor [CAR]
    • A61K2239/11Antigen recognition domain
    • A61K2239/13Antibody-based
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/10Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the structure of the chimeric antigen receptor [CAR]
    • A61K2239/17Hinge-spacer domain
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/55Lung
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the invention relates to the field of biomedical technology, in particular to a preparation method for obtaining a chimeric antigen receptor (CAR) exosomes by separation and an application thereof for treating diseases.
  • CAR chimeric antigen receptor
  • the first generation of CAR's genetic engineering technology consists of extracellular binding region-single-chain fragment variable (scFv), transmembrane region (TM) and intracellular signaling region-immunoreceptor tyrosine activating group.
  • composition of the chimeric antigen receptor CAR is linked as follows: svFv-TM-CD3 ⁇ (Zhang T, Barber A, Sentman C L. Chimeric NKG2D–Modified T Cells Inhibit Systemic T-Cell Lymphoma Growth in a Manner Involving Multiple Cytokines and Cytotoxic Pathways [J]. Cancer research, 2007, 67(22): 11029-11036.).
  • CD28 or CD137 also known as 4-1BB
  • the chimeric antigen receptor parts are connected as follows: scFv-TM- CD28-ITAM or scFv-TM-CD137-ITAM.
  • Co-stimulation of B7/CD28 or 4-1BBL/CD137 in the intracellular signal domain causes sustained proliferation of immune cells such as T cells, and can increase the level of cytokines such as IL-2 secreted by T cells (Savoldo B, et al. CD28)
  • Costimulation improves expansion and persistence of chimeric antigen receptor–modified T cells in lymphoma patients.
  • CAR cells have attractive prospects in clinical applications such as tumor immunotherapy.
  • problems such as the use of autologous immune cells, 1) it takes 10-14 days for the patient to complete the cell return after blood collection, which may miss the best time for patient treatment. 2) Patients often undergo multiple treatments such as radiotherapy and chemotherapy, and their physical condition is poor. The immune cells themselves have low activity and cannot guarantee the effectiveness of the transfused cells. 3) It may not be appropriate to collect blood from patients with advanced malignant diseases. 4) Large-scale application of immune cell reinfusion and massive proliferation of immune cells may cause inflammatory storms and become a dangerous clinical treatment complication. 5) The use of donor-derived CAR immune cells is likely to cause immune rejection.
  • immune cells can secrete a large number of exosomes, which are characterized by a diameter between 30-150 nm and a density between 1.13-1.19 g/mL; It carries important signaling molecules of immune cells, including proteins, lipids and RNA; it maintains similar life activities as the parental immune cells. It has certain cell killing potential when immune cells are activated.
  • Therapeutic potential of CAR-T cell-derived exosomes a cell-free modality for targeted cancer therapy.
  • the possibility of treating cancer with CAR-T extracellular bodies is proposed in the narrative.
  • the subsequent studies on the exosomes secreted by CAR-T cells are complex, have no specific targeting, and have no tangible tumor lethality.
  • Treatment of tumors with exosomes obtained by direct isolation of CAR-T cells No significant tumor suppressive effects were observed in in vivo experiments ( Figures 4-5). At present, no reports have been published on the effective treatment of exosome-derived diseases derived from CAR immune cells.
  • Exosomes produced by CAR-T cells face enormous challenges as a new method for cell-free treatment of diseases.
  • the inventors conducted intensive studies on exosomes secreted by CAR immune cells, and found that although CAR immunocytes secrete tumors have poor therapeutic effects, if specific antigens are used to stimulate CAR-T cells, they activate specific antigens.
  • the secreted exosomes are purified and enriched to prepare a specific exosome carrying CAR protein.
  • This exosome has a very strong anti-tumor effect.
  • it can be further engineered, such as encapsulating toxins, encapsulating radiation particles, and the like. It can treat diseases such as tumors.
  • the object of the present invention is to provide a chimeric antigen receptor (CAR) immune cell-derived CAR-containing immune cell exosomes (hereinafter referred to as "CAR exosomes”) preparation method and therapeutic disease thereof Applications.
  • CAR chimeric antigen receptor
  • CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia, 2014, 28(4): 917-927.
  • the preparation method employed in the present invention is not substantially different from the above method, and CAR immune cells prepared by the above-mentioned documents and general bioengineering means can be used in the present invention.
  • the immune cells may be T cells, NK cells or the like.
  • the source of immune cells can be the patient itself, or a healthy volunteer.
  • the immune cells are T cells derived from healthy volunteers and are carried out according to the following steps:
  • the immune cells are NK cells, scFv-hinge-TM-CD28-CD3 is constructed, a recombinant plasmid is produced and the virus is packaged to infect NK cells; and the CAR-NK cell population is expanded in vitro.
  • the immune cell is a CAR-T cell and a viral vector of scFv-hinge-CD28-4-1BB-CD3 is constructed.
  • the activator used may be a soluble recombinant protein of a specific target, an engineered cell expressing a specific target, or a tumor cell which expresses a specific target directly or the like.
  • the specific target refers to an antigen target targeted by scFv in CAR immune cells, that is, a specific target targeted by CAR immune cells.
  • the immobilized soluble recombinant protein antigen can achieve better activation effects than soluble recombinant protein antigens, such as magnetic beads coated with recombinant antigen.
  • activators derived from living cells often require inactivation.
  • the antigen target targeted by scFv in CAR immune cells may be a commonly used target for current targeted therapy such as EGFR, HER2, CD20, etc.
  • EGFR et al.
  • HER2 et al.
  • Current targeted therapy such as EGFR, HER2, CD20, etc.
  • the CAR immune cells used are CAR-T cells. Its target for scFv is EGFR.
  • the CAR immune cells used are CAR-NK cells. Its scFv targets the target HER2.
  • the activation method may be the addition of antigenic protein or solidified antigen protein in an in vitro culture system, direct co-cultivation of CAR immune cells, and inactivation of engineered cells expressing specific targets, direct co-cultivation of CAR immune cells, and expression specificity. Tumor cells, etc. after inactivation of the target.
  • the activator may specifically be: epidermal growth factor EGFR extracellular domain recombinant protein, magnetic bead cross-linked EGFR extracellular domain recombinant protein, EGFR-expressing CHO cells, EGFR-expressing MDA-MB-231 cells; magnetic beading The HER2 extracellular segment recombinant protein, HER2 expressing BT474 cells, and the like.
  • the activator is a magnetic bead coupled EGFR extracellular segment recombinant protein or an inactivated MDA-MB-231 cell with high expression of EGFR.
  • the activation method is specifically: culturing CAR-T cells in a medium containing a magnetic bead-coupled EGFR extracellular fusion protein or inactivating MDA-MB-231 with high expression of EGFR The cells are co-cultured.
  • the activator is a magnetic bead coupled HER2 extracellular segment recombinant protein or a HER2 overexpressing BT474 cell.
  • the activation method is specifically: the CAR-NK cells are cultured in a medium containing a magnetic bead-conjugated HER2 extracellular segment fusion protein or co-cultured with inactivated BT474 cells highly expressing HER2.
  • exosomes of CAR are basically not obtained by skipping this step B directly. That is, exosomes are directly obtained without activation of CAR immune cells by specific antigens, and then isolated and purified.
  • the purified CAR exosomes obtained in this way are very small and are basically not used for disease treatment or scientific research. However, it cannot be excluded that the method of large-scale cultivation can be used for enrichment and purification, but there is no application value from the economical consideration.
  • the culture supernatant was collected according to the activation method. Collected according to the general exosome separation method. Methods for collection of exosomes are mentioned in various literatures, such as Théry C, et al. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current protocols in cell biology, 2006: 3.22.1-3.22.29; Coumans F A W, et al. Methodological Guidelines to Study Extracellular Vesicles. Circulation research, 2017, 120(10): 1632-1648. Li L, et al. Human bile contains MicroRNA ⁇ laden extracellular vesicles that can be used for cholangiocarcinoma diagnosis Hepatology, 2014, 60(3): 896-907.
  • the exosomes are prepared as follows: the collected culture supernatant is centrifuged at 2000 g for 10 minutes at 4 ° C to remove dead cells and large debris; the supernatant is carefully treated Transfer to a new sterile centrifuge tube, centrifuge at 10000 g for 30 minutes at 4 ° C to remove organelles and small particles; carefully transfer the supernatant to a sterile ultracentrifuge tube and centrifuge at 110000g for 70 minutes at 4 ° C. The supernatant was carefully discarded, washed once with PBS, and ultracentrifuged at 110000 g for 70 minutes at 4 ° C, and the resulting precipitate was an exosomes.
  • the specific binding ability of the CAR receptor to the antigen is utilized to purify and enrich the CAR exosomes.
  • the binding of the protein L to the immunoglobulin light chain can be utilized to perform preliminary purification of exosomes secreted by the immune cells. It is worth noting that this method is not a substitute for the step of purifying the antigen.
  • the steps can be as follows:
  • a specific antigen-coated magnetic body that is, a CAR capture magnet containing a recombinant target protein antigen capable of CAR; and a resuspension after incubation
  • the magnetic field after removing the supernatant, adding a buffer, adding a sorting column, and eluting the exosomes retained on the sorting column with a buffer; after adding the supernatant to the sorting column, the first is the antigen-free binding.
  • the exosomes of the ability then elute the sorting column with a buffer, and the exudates are exosomes with antigen binding ability and carrying CAR.
  • PBS was resuspended as appropriate, total protein concentration was measured using a Bradford kit, and stored at -80 °C.
  • the magnetic beads coated with EGFR recombinant protein are used, the magnetic beads of which are Dynabeads.
  • the magnetic beads are added to the resuspension containing the exosomes, the test tube is placed in a magnetic field, and the exosomes specifically bound to the magnetic beads are adsorbed by the magnetic field; after the magnetic beads are adsorbed, the supernatant is aspirated, and the test tube is removed from the magnetic field.
  • Resuspend in suspension with PBS buffer add to the sorting column, collect the unbound components that flowed out first, and rinse the sorting column with buffer to wash out the exosomal vesicles without antigen binding ability.
  • the sorting column was removed from the magnetic field, and the exosomes retained on the sorting column were quickly eluted with buffer and equilibrated to physiological pH, at which time CAR exosomes were obtained.
  • the test tube is placed in a magnetic field, and the exosomes bound to the magnetic beads are adsorbed by the magnetic field; after the magnetic beads are adsorbed, the supernatant is aspirated, the test tube is removed from the magnetic field, suspended in suspension with PBS buffer, added to the sorting column, and collected.
  • the unbound component that has flowed out first, and the sorting column is washed with a buffer, and the exosomal vesicles that do not carry the immunoglobulin are washed off.
  • the sorting column is removed from the magnetic field, and the exosomes retained on the sorting column are quickly eluted with a buffer.
  • the pH was restored and the recombinant HER2 protein-coated magnetic beads were incubated at 4 ° C for 30 min; the supernatant was placed in a magnetic field.
  • the exosomes bound to the magnetic beads are again adsorbed by the magnetic field; after the magnetic beads are adsorbed, the supernatant is aspirated, the test tube is removed from the magnetic field, suspended in suspension with PBS buffer, added to the sorting column, and the unbound group that flows out first is collected.
  • the buffer was washed with a buffer and washed out with exosomal vesicles that did not carry CAR.
  • the sorting column is removed from the magnetic field, and the CAR-extracting exosomes retained on the sorting column are quickly eluted with a buffer and equilibrated to a physiological pH, and the eluted one is the target exosomes.
  • the invention detects the biological activity of the above CAR exosomes, and the exosomes carry CAR protein, and the average diameter is about 30-150 nm, and the morphology observed under the transmission electron microscope is consistent with the characteristics of the exosomes. Further studies have shown that the above-mentioned CAR-exposed immunocellular exosomes can well target cells and tissues expressed by the target, and can inhibit tumor cell proliferation and tumor growth in vivo.
  • a CAR exosome as described above and a composition thereof for the preparation of an antitumor medicament.
  • the tumors referred to in the present invention include adenocarcinoma, leukemia, lymphoma, melanoma, sarcoma, and the sources of tumor tissues include, but are not limited to, adrenal gland, gallbladder, bone, bone marrow, brain, breast, bile duct, gastrointestinal tract, heart, kidney. , liver, lungs, muscles, ovaries, pancreas, parathyroid glands, penis, prostate, skin, salivary glands, spleen, testes, thymus, thyroid, and uterus. In addition to the above tumors, it can also be used for tumors of the central nervous system such as glial cell tumors, astrocytomas, etc.
  • ocular tumors include basal cell carcinoma, squamous cell carcinoma, melanoma, etc., and also include endocrine glands. Tumors, neuroendocrine system tumors, gastrointestinal pancreatic endocrine system tumors, reproductive system tumors and head and neck tumors. It is not listed here one by one.
  • the tumor is non-small cell lung cancer or breast cancer.
  • the CAR exosomes inhibit cell viability and tumor growth rate of MDA-MB-231 and HCC827, particularly MDA-MB-231, which is naturally resistant to cetuximab.
  • the CAR exosomes inhibit the cellular viability of BT474.
  • the CAR exosomes are nano-membrane vesicles, which can be further combined with liposome-related engineering methods, or encapsulated with chemotherapeutic drugs, radioactive ions, and the like.
  • the CAR exosomes are further engineered for loading doxorubicin chemotherapeutic drugs and exhibit cytotoxic effects on breast cancer cells.
  • the anti-tumor drug referred to in the present invention refers to a drug having an inhibitory and/or therapeutic tumor, which may include a delay accompanying the development of symptoms associated with tumor growth and/or a decrease in the severity of these symptoms, and further includes an accompanying tumor growth accompanying The relief of symptoms and the prevention of other symptoms also reduce or prevent metastasis.
  • CAR exosomes are combined with an anti-tumor drug.
  • the CAR exosomes and compositions thereof disclosed in the present invention can also be combined with other anti-tumor drugs or radiotherapy for the treatment of tumors, and the anti-tumor drugs include: 1. Cytotoxic drugs (1) Drugs acting on the chemical structure of DNA: alkylating agents such as nitrogen mustards, nitrosouras, mesylate; platinum compounds such as cisplatin, carboplatin and platinum oxalate; mitomycin (MMC) (2) Drugs that affect nucleic acid synthesis: dihydrofolate reductase inhibitors such as methotrexate (MTX) and Alimta, etc; thymidine synthase inhibitors such as fluorouracil (5FU, FT-207, capecitabine) ⁇ nucleoside synthase inhibitors such as 6-mercaptopurine (6-MP) and 6-TG; nucleoside reductase inhibitors such as hydroxyurea (HU); DNA polymerase inhibitors such as cytarabine (Ar
  • hormone antiestrogens Tamoxifen, droloxifene, exemestane, etc.
  • aromatase inhibitors aminoglutethimide, lantron, letrozole, ruined, etc.
  • antiandrogen fluramide RH-LH agonist / antagonist Agents: Norred, enadine, etc.; 3, biological response modifiers: mainly through the body's immune function to inhibit tumor interferon; interleukin-2; thymosin; 4, monoclonal antibody: MabThera; Ma Sai Trastuzumab; Bevacizumab (Avastin); 5, various radiation therapy; 6, other including some drugs with unknown mechanism and further research; cell differentiation inducers such as retinoids; apoptosis inducers.
  • the CAR-containing immunocellular exosomes and compositions thereof disclosed herein can be administered in combination with one or a combination of the above-described antitumor drugs.
  • a CAR exosome as described above and a composition thereof for the preparation of a medicament for treating a severe infectious disease or an autoimmune disease.
  • a formulation which is a composition comprising a CAR exosome as described above.
  • the preparation is a composition comprising CAR exosomes, and the anti-tumor effect is remarkable after injection or other administration to an animal including a human. Specifically, it is effective for the prevention and/or treatment of tumors and can be used as an antitumor drug. In addition, due to the nature of immune cells, the exosomes and exosome compositions can also be used against other diseases, such as severe infectious diseases and autoimmune diseases.
  • a method of prolonging preparation for receiving, receiving or having received cancer treatment by administering to a patient a therapeutically effective amount of CAR exosomes eg, using chemotherapy, radiation therapy, targeted therapy, and/or surgery, etc.
  • a therapeutically effective amount of CAR exosomes eg, using chemotherapy, radiation therapy, targeted therapy, and/or surgery, etc.
  • a method of recurrence-free survival in cancer patients Due to the tissue penetrability of exosomes, it should be more advantageous in the treatment of solid tumors than the treatment of CAR immune cells.
  • the dosages to be administered vary depending on the age and weight of the patient, the disease characteristics and severity, and the route of administration, and may be referred to animal experiments. The results and various conditions, the total dose can not exceed a certain range.
  • the resulting exosomes are further analyzed, isolated, purified, and enriched, and finally the immune cell exosomes carrying the CAR are obtained.
  • the exosomes can be used for the treatment of various diseases, such as cancer, severe infectious diseases, etc., and the exosomes have the ability to overcome adverse reactions such as immune inflammatory storms treated by CAR cells, and enhance the tissue soaking ability of the CAR. It has the advantages of convenient storage and transportation, and provides a new strategy for the treatment of related diseases.
  • the invention is further illustrated by the following examples and experimental examples, which are not to be construed as limiting the invention.
  • the examples do not include detailed descriptions of conventional methods, such as those used to construct vectors and plasmids, methods of inserting genes encoding proteins into such vectors and plasmids, or methods of introducing plasmids into host cells, or methods of packaging viruses A method of infecting immune cells with a virus to obtain expression of a protein of interest.
  • Such methods are well known to those of ordinary skill in the art and are described in a number of publications, including Sambrook, J., Fritsch, EF and Maniais, T.
  • CAR sequence comprising an anti-EGFR single-chain antibody (commissioned by Suzhou Jinweizhi Biotechnology Co., Ltd.), wherein the single-chain antibody sequence is derived from the anti-EGFR antibody cetuximab (Li et al., 2005, Structural basis for inhibition of
  • the specific structure of CAR includes: anti-EGFR single-chain antibody scFv-CD8 ⁇ hinge region and transmembrane region-4-1BB co-activation domain and CD3 ⁇ signaling molecule Intracellular segment.
  • the specific sequence is generally identical to that reported in the literature by Johnson L A, et al. Science translational medicine, 2015, 7 (275) (except scFv).
  • the Myc tag is inserted between the scFv and the hinge region, and the position of the tag is related to the literature.
  • Chu J, et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human Multiple myeloma. Leukemia, 2014, 28(4): 917-927. Same. The entire sequence was cloned into the pLenti6.3/v5 lentiviral vector (Invitrogen) containing the CMV promoter by homologous recombination.
  • PBMC peripheral blood mononuclear cells
  • paramagnetic beads Distynabeads ClinExVivo CD3/CD28, Invitrogen, Camarillo, CA, USA
  • anti-CD3 and anti-CD28 antibodies were used. Stimulate and enrich, the ratio of magnetic beads to cells is 2-3:1; the cells are diluted to a concentration of 5-8 ⁇ 10 6 /mL, and cultured for 24 hours in the medium supplemented with IL-2; the obtained T cells are slow
  • the virus is infected multiple times.
  • the cells were counted every other day and the medium was changed.
  • the next experiment was performed when the T cells showed a static state. By resting state is meant that the proliferation coefficient decreases in cell count and the cell size stops changing. At this time, the supernatant of the cells before and after transfection was collected to extract exosomes for use as a control. The method of extraction will be described later.
  • T cell specific antigen activation In this step, two methods are used for CAR-T cell-specific antigen activation.
  • the first method is to add a magnetic bead-conjugated EGFR extracellular domain recombinant protein to the T cell culture medium. The experiment was carried out with a protein concentration ranging from 5 ug/mL to 1 mg/mL. The culture supernatant was collected at 24 hours. The recombinant protein in the supernatant is removed by a magnetic field.
  • the second method is co-cultivation of CAR-T cells with tumor cells MDA-MB-231 with high expression of EGFR. MDA-MB-231 cells were inactivated with 100 Gy of gamma rays prior to incubation. The incubation ratios were 2:1, 4:1 and 8:1, and the culture supernatant was collected 24 hours later.
  • Exosomes preparation The culture supernatants described in 3) and 4) were extracted as follows: The culture supernatant was placed in a 500 ml sterile centrifuge bottle or a 50 ml polypropylene centrifuge tube (purchased from Beckman) at 4 ° C, 2000 g. Centrifuge for 10 minutes to remove dead cells and large debris. The supernatant was carefully transferred to a new sterile centrifuge tube and centrifuged at 10,000 g for 30 minutes at 4 ° C to remove organelles and small particles.
  • the obtained exosomes were purified and enriched with CAR-extracting exosomes according to the following steps: magnetic beads coated with EGFR extracellular domain recombinant protein were added to physiological saline solution containing exosomes, and incubated at 4 ° C for 30 min, magnetic beads The specific antigen-antibody interaction is combined with the CAR exosomes with the corresponding svFv; the test tube is placed in a magnetic field, and the exosomes connected to the magnetic beads are adsorbed by the magnetic field; after the magnetic beads are adsorbed, the supernatant is aspirated, The test tube was removed from the magnetic field; resuspended in suspension with PBS buffer, added to a sorting column, and the unbound components that flowed out first were collected, and the sorting column was washed with a buffer, and the exosomal vesicles that did not carry CAR were washed off.
  • the sorting column is removed from the magnetic field, and the CAR-extracting exosomes retained on the sorting column are quickly eluted with a buffer, and the eluted exudates are the target exosomes.
  • the TBST was washed 3 times and then HRP-labeled Anti-Myc antibody was added. Incubate for 1 h at 37 °C. After washing with TBST, a chromogenic substrate was added. After the microplate reader was tested and calculated, the results are shown in Figure 1.
  • the competition ELISA with Cetuximab was as follows: CAR exosomes and biotin-labeled Cetuximab were added to a well-sealed 96-well plate coated with EGFR antigen at a certain dilution ratio, and incubated at 37 ° C for 1 h. The TBST was washed 3 times, followed by the addition of HRP-labeled avidin (Thermo). Incubate for 1 h at 37 °C. After washing with TBST, a chromogenic substrate was added. After the detection of the microplate reader, the calculation and analysis results are shown in Fig. 2.
  • the morphology of the CAR exosomes obtained by transmission electron microscopy was observed: the exosomes were fully resuspended, and 10 ⁇ l of the extract was pipetted onto the loaded copper mesh, allowed to stand at room temperature for 5 minutes, and the excess liquid was carefully sucked off with a filter paper. Negative dyeing of uranyl acetate was added dropwise for 2 minutes, excess liquid was sucked off with filter paper, and dried under an incandescent lamp; 80-120 kv was imaged by a transmission electron microscope and photographed. As a result, as shown in the figure, a circular vesicular structure having a diameter of about 30 to 150 nm was observed, and the results are shown in Fig. 3.
  • the results of this example demonstrate that the CAR-derived exosomes derived from immune cells are successfully obtained. They all express CAR protein and bind to specific antigens. The average diameter is about 80 nm. The morphology observed under transmission electron microscope is consistent with exocrine. Body characteristics.
  • Example 2 CAR exosomes inhibit EGFR-positive MDA-MB-231 and HCC827 cell viability experiments
  • MDA-MB-231 and HCC827 cells (ATCC) with good growth conditions were adjusted to a cell concentration of 5 ⁇ 10 3 /ml, seeded in 96-well cell culture plates, 200 ⁇ l/well, and incubated at 37 ° C, 5% CO 2 After 24 hours of culture, a final concentration of 5 nmol of EGF and different concentrations of exosomes were added to the culture medium, and cetuximab antibody was used as a control (Cetuximab was purchased from Merck). After 4 days, cell viability was used with CellTiter-Glo. Luminescent Cell Viability Assay Kit (Promega, Madison, WI). The experimental results are shown in Figure 3. The results showed that CAR exosomes significantly inhibited MDA-MB-231 and HCC827 cell viability (P ⁇ 0.01, Tukey's test), especially for Cetuximab-resistant MDA-MB-231 cells. ( Figure 4).
  • Example 3 Inhibition of tumor growth in CAR exosomes in vivo
  • HCC827 and MDA-231 cells were first inoculated into the right flank of BALB/c nude mice (Chinese Academy of Experimental Animals Center), and 3500 mg/kg CAR was injected into the tail vein after tumor formation.
  • the exosomes and the antibody drug cetuximab (10 mg/kg) were injected once a week until the tumor was over-exposed. The length and width of the tumor were measured daily to calculate the tumor volume.
  • the tumor growth curve is shown in Figure 5. The results showed that the tumor growth rate of the activated CAR exosomes treated group was significantly lower than that of the cetuximab treatment group (P ⁇ 0.01 after 40 days, Bonferroni test).
  • CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma.
  • the entire sequence was cloned into the PCDH lentiviral vector (System Biosciences) containing the CMV promoter by homologous recombination.
  • NK-92 cells were diluted to 1 ⁇ 10 6 /mL and cultured overnight in medium supplemented with IL-2, followed by repeated infection with lentivirus. After the third infection, the cells were cultured in 1640 medium containing 20% FBS. This medium was supplemented with 150 units/ml of IL-2. Green fluorescent protein expressed cells were sorted by flow cytometry (BD Biosciences, San Jose, CA, USA). The green fluorescent protein is encoded by the PCDH vector gene. In addition, cell supernatants before and after infection were collected in the experiment to extract exosomes for use as a control. The method of extraction will be described later.
  • NK cell specific antigen activation In this step, two methods are used for NK cell-specific antigen activation.
  • the first method is to add a magnetic bead-conjugated HER2 extracellular segment recombinant protein to the NK cell culture medium. The experiment was carried out with a protein concentration ranging from 5 ug/mL to 1 mg/mL. The culture supernatant was collected at 12-24 hours.
  • the second method is co-culture of NK cells with inactivated BT474 cells with high expression of HER2. The incubation ratios were 2:1, 4:1 and 8:1, and the culture supernatant was collected after 12-24 hours.
  • Exosomes preparation The culture supernatants described in 3) and 4) were subjected to extraction of exosomes as follows: The culture supernatant was placed in a 500 ml sterile centrifuge bottle or a 50 ml polypropylene centrifuge tube (purchased from Beckman) at 4 ° C, 2000 g. Centrifuge for 10 minutes to remove dead cells and large debris. The supernatant was carefully transferred to a new sterile centrifuge tube and centrifuged at 10,000 g for 30 minutes at 4 ° C to remove organelles and small particles.
  • the obtained exosomes are purified and enriched with CAR-extracting exosomes according to the following steps: First, magnetic beads coated with protein L are added to a physiological saline solution containing exosomes, and incubated at 4 ° C for 60 min, and the magnetic beads pass through protein L.
  • the sorting column was removed from the magnetic field, and the exosomes retained on the sorting column were quickly eluted with buffer and immediately incubated with the recombinant HER2 protein-coated magnetic beads at 4 ° C for 30 min.
  • the magnetic beads were passed through specific antigen-antibody interactions.
  • the sorting column is removed from the magnetic field, and the CAR-extracting exosomes retained on the sorting column are quickly eluted with a buffer and equilibrated to a physiological pH, and the eluted one is the target exosomes. After detecting the total protein concentration using the Bradford kit (purchased from Thermo), it can be stored at -80 ° C for long-term storage.
  • the morphology of the CAR exosomes obtained by transmission electron microscopy was observed: the exosomes were fully resuspended, and 10 ul of the extract was pipetted onto the loaded copper mesh, allowed to stand at room temperature for 5 minutes, and the excess liquid was carefully sucked off with a filter paper. Negative dyeing of uranyl acetate was added dropwise for 2 minutes, excess liquid was sucked off with filter paper, and dried under an incandescent lamp; 80-120 kv was imaged by a transmission electron microscope and photographed. As a result, as shown in the figure, a circular vesicular structure having a diameter of about 30 to 150 nm was observed, and the results are shown in Fig. 6.
  • Example 5 NK-derived CAR exosomes inhibit breast cancer cell viability assay
  • the HER2 high-expressing breast cancer cell line BT474 and HER2 low-expressing MCF-7 cells (ATCC) with good growth status were adjusted to a cell concentration of 4 ⁇ 10 3 /ml, and seeded in a 96-well cell culture plate at 200 ⁇ l/well. After incubation for 24 h in a °C, 5% CO 2 incubator, exosomes of different concentration gradients were added to the culture medium, and trastuzumab antibody drug was used as a control. After 4 days, the CellTiter-Glo Luminescent Cell Viability Assay kit was used for cell viability ( Promega, Madison, WI) testing. The experimental results are shown in Figure 7.
  • Example 6 Preparation of NK-derived CAR exosomes loaded with doxorubicin and their antitumor effects
  • the CAR exosomes obtained in Examples 4 and 5 were mixed with an equal mass of doxorubicin chemotherapeutic drug.
  • Compound loaded CAR exosomes were prepared by electroporation.
  • the electric shock condition was a voltage of 420 V and a capacitance of 150 ⁇ F, which was electrically rotated in a 4 mm electric rotor. Subsequently, it was filtered through an inverted centrifugal ultrafiltration membrane to remove free compounds that were not transfected into the exosomes.
  • the exosomes can also be loaded by introducing the compounds into the exosomes by lipofection.
  • the HER2 high-expressing breast cancer cell line BT474 and HER2 low-expressing MCF-7 cells (ATCC) with good growth status were adjusted to a cell concentration of 4 ⁇ 10 3 /ml, and seeded in a 96-well cell culture plate at 200 ⁇ l/well. After incubation for 24 h in °C and 5% CO 2 incubator, exosomes of different concentration gradients were added to the culture medium. After 4 days, cell viability was detected by CellTiter-Glo Luminescent Cell Viability Assay Kit (Promega, Madison, WI). .
  • the experimental results are shown in Figure 7. The results showed that CAR excipients loaded with doxorubicin could significantly inhibit tumor cell viability (P ⁇ 0.01, Tukey test).
  • Example 7 Effect of CAR-T-derived CAR exosomes targeting CD20 on lymphoma cell apoptosis.
  • CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma.
  • the entire sequence was cloned into the PCDH lentiviral vector (System Biosciences) containing the CMV promoter by homologous recombination.
  • the method for preparing lentivirus using HEK293T cells, the method for preparing CAR-T cells, the method for activating stimulation of CAR-T cell-specific antigen, the preparation of exosomes, and the purification method for carrying CAR exosomes are the same as the above examples, No longer.
  • the specific antigen activator of CAR-T cells is an inactivated Raji cell expressing CD20.
  • the Burkitt lymphoma cell line Raji (ATCC), which grew well, was adjusted to a cell concentration of 1 ⁇ 10 5 /well. After incubating for 24 hours at 37 ° C in a 5% CO 2 incubator, exosomes of different concentration gradients were added to the culture medium, and Rituximab antibody drug was used as a control. The cells were rinsed for 16 hours, and then annexin V was used. After FITC (BD Biosciences) staining, flow cytometry was performed to calculate the ratio of apoptotic cells. The experimental results are shown in Figure 8. The results showed that CAR-T cells derived from CAR exosomes can significantly induce apoptosis in Raji cells and Daudi cells (P ⁇ 0.01, Tukey test). ( Figure 8).

Landscapes

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

Abstract

提供一种利用分离获得携带CAR的免疫细胞外泌体的制备方法及其应用。将CAR免疫细胞以特异性的抗原活化后,产生的外泌体进行进一步的分析、分离、纯化、富集,最终得到携带CAR的免疫细胞外泌体。该外泌体能用于癌症,重型传染病等疾病的治疗。

Description

携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用 技术领域
本发明涉及生物医药技术领域,具体地说,是一种利用分离获得含嵌合抗原受体(Chimeric Antigen Receptor,CAR)外泌体的制备方法及其治疗疾病的应用。
背景技术
以手术治疗、放射治疗及化学治疗为主,辅以新型的靶向治疗方案是近年来对恶性肿瘤治疗的基本策略,且已经在临床实践中取得重要的进展。但恶性肿瘤的复发、转移和治疗性耐受依然是一直困扰临床和科研工作者的难题。近年来,基因修饰各种免疫细胞用于治疗疾病的方法已经提出,如通过嵌合抗原受体(Chimeric Antigen Receptor,CAR)在T细胞上的表达,使得基因修饰的T细胞靶向肿瘤细胞上表达的抗原,以治疗癌症类疾病。这类的治疗方式已经获得一定程度上的成功,第一个类似的产品也与2017年或FDA批准(Brentjens R,et al.Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells:case report of an unforeseen adverse event in a phase I clinical trial,Molecular Therapy,2010,18(4):666-668.)随着目前技术的发展,目前嵌合抗原受体细胞技术中嵌合抗原受体基因构建的方式主要划分以下三代。第一代CAR的基因工程技术由胞外结合区-单链抗体(Single-chain fragment variable,scFv)、跨膜区(Transmembrane Region,TM)和胞内信号区-免疫受体酪氨酸活化基序(immuno-receptor tyrosine-based activation motif,ITAM)组成,其中嵌合抗原受体CAR部分按照如下形式连接:svFv-TM-CD3ζ(Zhang T,Barber A,Sentman C L.Chimeric NKG2D–Modified T Cells Inhibit Systemic T-Cell Lymphoma Growth in a Manner Involving Multiple Cytokines and Cytotoxic Pathways[J].Cancer research,2007,67(22):11029-11036.)。随后发展的第二代CAR基因工程策略在一代的基础上增加了CD28或CD137(又名4-1BB)的胞内信号区,其中嵌合抗原受体各部分按照如下形式连接:scFv-TM-CD28-ITAM或scFv-TM-CD137-ITAM。胞内信号区发生的B7/CD28或4-1BBL/CD137共刺激作用引起T细胞等免疫细胞的持续增殖,并能够提高T细胞分泌IL-2等细胞因子的水平(Savoldo B,et al.CD28 costimulation improves expansion and  persistence of chimeric antigen receptor–modified T cells in lymphoma patients.The Journal of clinical investigation,2011,121(5):1822.)。近年来发展的第三代CAR基因工程连接如下:scFv-TM-CD28-CD137-ITAM或scFv-TM-28-CD134-ITAM,进一步提高CAR-T在体内的存活周期和效果(Carpenito C,et al.Control of large,established tumor xenografts with genetically retargeted human T cells containing CD28and CD137domains.Proceedings of the National Academy of Sciences,2009,106(9):3360-3365.)。除了最常用的T细胞,近年来也涌现出利用CAR技术制备其他免疫细胞类型的治疗方法,如CAR-NK[Chu J,et al.CS1-specific chimeric antigen receptor(CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma.Leukemia,2014,28(4):917-927.]等。
CAR细胞在肿瘤免疫治疗等临床应用中具有诱人的前景。但目前有如下的显著问题:如利用自体的免疫细胞,1)患者采血后需要10-14天才能完成细胞回输,这段时间可能错过病人治疗的最佳时机。2)患者往往经过多重治疗如放化疗等,身体状况差,免疫细胞本身活性低,无法保证回输细胞的有效性。3)晚期恶性疾病病人采集血液可能不适宜。4)大规模应用免疫细胞回输及免疫细胞的大量增殖有可能造成炎性风暴,成为凶险的临床治疗并发症。5)而利用供体来源的CAR免疫细胞容易造成免疫排斥反应。
值得关注的是,免疫细胞能分泌大量的外泌体(exosome),这些外泌体的特点是:直径在30-150nm之间;密度在1.13-1.19g/mL之间;表达特异性的蛋白,携带了免疫细胞的重要信号分子,包括蛋白质、脂质和RNA等;保持着与亲代免疫细胞相似的生活学活性。在免疫细胞激活时,具有一定的细胞杀伤潜能。
非专利文献综述Tang X J,et al.Therapeutic potential of CAR-T cell-derived exosomes:a cell-free modality for targeted cancer therapy.Oncotarget,2015,6(42):44179。在叙述中提出利用CAR-T细胞外泌体治疗癌症的可能性。但后续的研究CAR-T细胞分泌的外泌体成分复杂,无特异靶向性,也无切实的肿瘤杀伤力。使用直接分离获得CAR-T细胞的外泌体治疗肿瘤在体内实验中并未观察到明显的肿瘤抑制效果(附图4-5)。且目前尚未见到任何CAR免疫细胞来源的外泌体治疗疾病有效的报道。CAR-T细胞产生的外泌体作为无细胞治疗疾病的新方法又面临巨大挑战。
最近,发明人针对CAR免疫细胞所分泌的外泌体成分进行了深入研究,结果发现虽然CAR免疫细胞分泌体肿瘤治疗效果差,如果使用特定抗原刺激CAR-T细胞,使其对特定的抗原活化,再对其分泌的外泌体进行纯化、富集,以制备特定的携带有CAR蛋白的外泌体。该种外泌体有非常强的抗肿瘤作用。且由于外泌体的膜性结构,可以进一步对其进行工程改造,如包裹毒素、包裹放射粒子等。可以实现肿瘤等疾病的治疗。
公开于该背景技术部分的信息仅仅旨在增加对本发明的总体背景的理解,而不应当被视为承认或以任何形式暗示该信息构成已为本领域一般技术人员所公知的现有技术。
发明内容
本发明的目的在于提供一种嵌合抗原受体(Chimeric Antigen Receptor,CAR)免疫细胞来源的携带CAR的免疫细胞外泌体(后述为“CAR外泌体”)的制备方法及其治疗疾病的应用。
本发明的第一方面,提供上述的CAR外泌体的制备方法,包括以下步骤:
A)制备CAR免疫细胞
需要说明的是,本步骤所述的CAR免疫细胞的制备方法在多种文献中提及,如Johnson L A,,et al.Rational development and characterization of humanized anti–EGFR variant III chimeric antigen receptor T cells for glioblastoma.Science translational medicine,2015,7(275):275ra22-275ra22;Park S,et al.Micromolar affinity CAR T cells to ICAM-1 achieves rapid tumor elimination while avoiding systemic toxicity.Scientific reports,2017,7(1):14366.;Li N,et al.Therapeutically targeting glypican-2 via single-domain antibody-based chimeric antigen receptors and immunotoxins in neuroblastoma Proceedings of the National Academy of Sciences,2017,114(32):E6623-E6631.Chu J,et al.CS1-specific chimeric antigen receptor(CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma.Leukemia,2014,28(4):917-927.等方法。本发明采用的制备方法和上述方法并无本质区别,利用上述文献中报道的以及通用的生物工程手段制备的CAR免疫细胞均可以用于本发明中。其中免疫细胞可以是T细胞、NK细胞等。免疫细胞的来源可以是患者自身,或健康志愿者。
在本发明的一个具体实施例中,免疫细胞为健康志愿者来源的T细胞,按照以下步骤实施:
(1)获得细胞样本并分离和激活,所述样本为T细胞或T细胞的祖细胞;
(2)构建scFv-CD8hinge and TM-4-1BB-CD3的病毒载体;
(3)制作重组质粒并包装病毒;
(4)病毒感染T细胞;
(5)体外培养扩增CAR-T细胞群体。
在本发明的另一个具体实施例中,免疫细胞为NK细胞,构建scFv-hinge-TM-CD28-CD3,制作重组质粒并包装病毒后感染NK细胞;体外扩增CAR-NK细胞群体。在本发明的另一个具体实施例中,免疫细胞为CAR-T细胞,构建scFv-hinge-CD28-4-1BB-CD3的病毒载体。
B)对CAR免疫细胞进行抗原特异性活化:
获得大量CAR免疫细胞后,需进行CAR免疫细胞的抗原特异性活化。
在此步骤中,采用的活化剂可以是特异性靶点的可溶性重组蛋白、表达特异性靶点的工程细胞、或直接使用表达特异性靶点肿瘤细胞等。所述的特异性靶点指的是CAR免疫细胞中的scFv针对的抗原靶点,即CAR免疫细胞靶向的特异性靶点。值得注意的是,利用固定化的溶性重组蛋白抗原可以获得相比可溶性重组蛋白抗原更好的激活效果,如重组抗原包被的磁珠等。另一个注意要点是,来源于活细胞的活化剂常需要灭活处理。
CAR免疫细胞中的scFv针对的抗原靶点,可以是EGFR,HER2,CD20等目前靶向疗法中常用靶标[Caruso H G,et al.Tuning sensitivity of CAR to EGFR density limits recognition of normal tissue while maintaining potent antitumor activity[J].Cancer research,2015,75(17):3505-3518.][Ahmed N,et al.Human Epidermal Growth Factor Receptor 2(HER2)-Specific Chimeric Antigen Receptor–Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma.Journal of Clinical Oncology,2015,33(15):1688-1696.],也可以为CD19,Mesothelin等肿瘤抗原(Turtle C J,et al.CD19CAR–T cells of defined CD4+:CD8+composition in adult B cell ALL patients.The Journal of clinical investigation,2016,126(6):2123.)。原则上来讲,针对任何靶点的CAR免疫细胞均可以在本步骤中使用。
在本发明的一个具体实施例中,采用的CAR免疫细胞为CAR-T细胞。其 scFv针对的靶点为EGFR。
在本发明的另一个具体实施例中,采用的CAR免疫细胞为CAR-NK细胞。其scFv针对的靶点为HER2。
活化的方法可以为在体外的培养体系中加入抗原蛋白或固化的抗原蛋白、直接共培育CAR免疫细胞及和表达特异性靶点的灭活后工程细胞、直接共培育CAR免疫细胞和表达特异性靶点的灭活后肿瘤细胞等。
所述的活化剂具体可为:表皮生长因子EGFR胞外段重组蛋白、磁珠交联的EGFR胞外段重组蛋白,表达EGFR的CHO细胞、表达EGFR的MDA-MB-231细胞;磁珠交联的HER2胞外段重组蛋白、表达HER2的BT474细胞等。
在本发明的一个具体实施例中,所述的活化剂为磁珠偶联的EGFR胞外段重组蛋白或高表达EGFR的灭活MDA-MB-231细胞。在该实施例中,所述的活化方法具体为:将CAR-T细胞培养于含有磁珠偶联的EGFR胞外段融合蛋白的培养基中或与高表达EGFR的灭活MDA-MB-231细胞共培育。
在本发明的另一个具体的实施例中,所述的活化剂为磁珠偶联的HER2胞外段重组蛋白或HER2高表达的BT474细胞。所述的活化方法具体为:将CAR-NK细胞培养于含有磁珠偶联的HER2胞外段融合蛋白的培养基中或与高表达HER2的灭活后BT474细胞进行共培育。
值得说明的是,直接跳过本步骤B基本无法获得CAR的外泌体。即不经过特异性抗原活化CAR免疫细胞而直接获取外泌体,再进行分离纯化。这种方式纯化后得到的CAR外泌体含量十分少,基本无法用于疾病治疗或科学研究。但不排除可以应用大规模培养的方法富集纯化,但从经济性考虑并无应用价值。
C)收集制备CAR免疫细胞外泌体:
根据活化方式的不同,采集培养上清。按照通用的外泌体分离方法进行收集制备。外泌体收集的方法在多种文献中提及,如Théry C,et al.Isolation and characterization of exosomes from cell culture supernatants and biological fluids.Current protocols in cell biology,2006:3.22.1-3.22.29;Coumans F A W,et al.Methodological Guidelines to Study Extracellular Vesicles.Circulation research,2017,120(10):1632-1648.Li L,et al.Human bile contains MicroRNA‐laden extracellular vesicles that can be used for cholangiocarcinoma diagnosis. Hepatology,2014,60(3):896-907.Li L,Piontek K,Ishida M,et al.Extracellular vesicles carry microRNA‐195 to intrahepatic cholangiocarcinoma and improve survival in a rat model.Hepatology,2017,65(2):501-514.分离外泌体的技术手段在本步骤中与上述文献并无本质区别。
在本发明的一个具体实施例中,所述的外泌体按以下方法制备:将收集的培养上清于4℃、2000g离心10分钟,以去除死细胞和大的碎片;小心将上清液转移到新的无菌离心管中,于4℃、10000g离心30分钟,以去除细胞器及小颗粒;小心将上清液转移到无菌超速离心管中,于4℃,110000g超速离心70分钟,小心弃去上清,再加PBS清洗一次,于4℃,110000g超速离心70分钟,得到的沉淀即为外泌体。
D)CAR外泌体的纯化和富集:
在该步骤中,利用CAR受体与抗原的特异性结合能力以纯化和富集CAR外泌体。为了增加纯度,在某些具体应用实施例中可以利用蛋白质L与免疫球蛋白轻链的结合力对免疫细胞分泌的外泌体进行初步纯化。值得注意的是,该方法不能替代利用抗原纯化的步骤。步骤可以如下:
向步骤C中制得的外泌体重悬液中加入特异性抗原包被的磁性体,即CAR捕获磁体,所述磁性体上含有能够CAR的重组靶点蛋白抗原;孵育后将重悬液置于磁场中,除去上清液后加入缓冲液,加入分选柱,利用缓冲液洗脱分选柱上滞留的外泌体;将上清液加入分选柱后,先流出的是无抗原结合能力的外泌体,然后采用缓冲液洗脱分选柱,流出的即为具有抗原结合能力、携带CAR的外泌体。根据最初收集的培养基的体积不同,酌情加入PBS重悬,用Bradford试剂盒检测总蛋白浓度,-80℃分装保存。
在本发明的一个具体的实施例中,采用EGFR重组蛋白包被的磁珠,其磁珠为Dynabeads。向含有外泌体的重悬液加入该磁珠,将试管置于磁场中,与磁珠特异性结合的外泌体被磁场吸附;磁珠被吸附后,吸去上清,将试管移出磁场;用PBS缓冲液悬浮重悬,加入分选柱中,收集先行流出的未结合组分,并用缓冲液冲洗分选柱,洗下来的为无抗原结合能力的外泌体囊泡。将分选柱移出磁场,用缓冲液快速将分选柱上滞留的外泌体洗脱下来并平衡至生理pH,此时获得CAR外泌体。
在本发明的另一个具体实施例中,该步骤为首先利用包被有蛋白L的磁珠与含有外泌体的PBS溶液4℃孵育60min,磁珠通过蛋白L与免疫球蛋白轻链 的特异性结合力结合对应的外泌体。将试管置于磁场中,与磁珠结合的外泌体被磁场吸附;磁珠被吸附后,吸去上清,将试管移出磁场;用PBS缓冲液悬浮重悬,加入分选柱中,收集先行流出的未结合组分,并用缓冲液冲洗分选柱,洗下来的为不携带有免疫球蛋白的外泌体囊泡。将分选柱移出磁场,用缓冲液快速将分选柱上滞留的外泌体洗脱下来后。然后立即恢复pH值后与重组HER2蛋白包被的磁珠在4℃孵育30min;将上清液置于磁场中。与磁珠结合的外泌体再次被磁场吸附;磁珠被吸附后,吸去上清,将试管移出磁场;用PBS缓冲液悬浮重悬,加入分选柱中,收集先行流出的未结合组分,用缓冲液冲洗分选柱,洗下来的为不携带有CAR的外泌体囊泡。将分选柱移出磁场,用缓冲液快速将分选柱上滞留的携带有CAR的外泌体洗脱下来并平衡至生理pH,洗脱下来的即为目的外泌体。
本发明的第二方面,提供一种采用如上所述的制备方法制备得到的CAR外泌体。
本发明对上述CAR外泌体进行生物学活性的检测,所述的外泌体携带CAR蛋白,平均直径在30-150nm左右,透射电镜下观察到的形态都符合外泌体的特征。进一步的研究显示,上述携带CAR的免疫细胞外泌体可以很好地靶向目标靶点表达的细胞及组织,并且可以抑制肿瘤细胞增殖和体内肿瘤生长。
本发明的第三方面,提供一种如上所述的CAR外泌体及其组合物在制备抗肿瘤药物中的应用。
本发明所指的肿瘤,包括腺癌、白血病、淋巴瘤、黑色素瘤、肉瘤,肿瘤组织的来源包括但不限于肾上腺、胆囊、骨、骨髓、脑、乳腺、胆管、胃肠道、心脏、肾脏、肝脏、肺、肌肉、卵巢、胰腺、甲状旁腺、阴茎、前列腺、皮肤、唾液腺、脾脏、睾丸、胸腺、甲状腺和子宫。除了上述的肿瘤外,还可用于中枢神经系统的肿瘤如胶质细胞多样性瘤、星细胞瘤等,此外眼部的肿瘤包括基底细胞癌、鳞状细胞癌、黑色素瘤等,还包括内分泌腺肿瘤、神经内分泌系统肿瘤、胃肠道胰腺内分泌系统肿瘤,生殖系统肿瘤及头颈部肿瘤等。这里不再一一列举。
进一步的,所述的肿瘤为非小细胞肺癌或乳腺癌。在本发明的一个具体实施例中,所述的CAR外泌体抑制MDA-MB-231和HCC827的细胞活力和肿瘤 生长速度,尤其是对cetuximab天然耐药的MDA-MB-231。在本发明的另一个具体实施例中,所述的CAR外泌体抑制BT474的细胞活力。
进一步的,所述的CAR外泌体是一种纳米膜性囊泡,可以进一步和脂质体相关的工程改造方法结合,或包裹化疗药物、放射离子等。
在一个具体的实施例中,CAR外泌体进一步工程化用于负载阿霉素化疗药物并对乳腺癌细胞显示出细胞毒性作用。
本发明所称的抗肿瘤药物,指具有抑制和/或治疗肿瘤的药物,可以包括伴随肿瘤生长相关症状发展的延迟和/或这些症状严重程度的降低,它进一步还包括已存在的肿瘤生长伴随症状的减轻并防止其他症状的出现,还也减少或防止转移。
进一步的,所述的CAR外泌体和抗肿瘤药联用。
本发明公开的所述的CAR外泌体及其组合物还可以和其他的抗肿瘤药联合给药或放射治疗,用于肿瘤的治疗,这些抗肿瘤药包括:1、细胞毒类药物(1)作用于DNA化学结构的药物:烷化剂如氮芥类、亚硝尿类、甲基磺酸酯类;铂类化合物如顺铂、卡铂和草酸铂等;丝裂霉素(MMC);(2)影响核酸合成的药物:二氢叶酸还原酶抑制剂如甲氨喋呤(MTX)和Alimta等;胸腺核苷合成酶抑制剂如氟尿嘧啶类(5FU、FT-207、卡培他滨)等;嘌呤核苷合成酶抑制剂如6-巯基嘌呤(6-MP)和6-TG等;核苷酸还原酶抑制剂如羟基脲(HU)等;DNA多聚酶抑制剂如阿糖胞苷(Ara-C)和健择(Gemz)等;(3)作用于核酸转录的药物:选择性作用于DNA模板,抑制DNA依赖RNA聚合酶,从而抑制RNA合成的药物如:放线菌素D、柔红霉素、阿霉素、表阿霉素、阿克拉霉素、光辉霉素等;(4)主要作用于微管蛋白合成的药物:紫杉醇、泰索帝、长春花碱、长春瑞滨、鬼臼硷类、高三尖杉酯碱;(5)其他细胞毒药:门冬酰胺酶主要抑制蛋白质的合成;2、激素类抗雌激素:三苯氧胺、屈洛昔芬、依西美坦等;芳香化酶抑制剂:氨鲁米特、兰特隆、来曲唑、瑞宁德等;抗雄激素:氟它氨RH-LH激动剂/拮抗剂:诺雷德、依那通等;3、生物反应调节剂:主要通过机体免疫功能抑制肿瘤干扰素;白细胞介素-2;胸腺肽类;4、单克隆抗体:美罗华(MabThera);赫赛汀(Trastuzumab);Bevacizumab(Avastin);5、各种放射疗法;6、其他包括一些目前机制不明和有待进一步研究的药物;细胞分化诱导剂如维甲类;细胞凋亡诱导剂。本发明 公开的携带CAR的免疫细胞外泌体及其组合物可以和上述的抗肿瘤药物之一或其组合联合用药。
本发明的第四方面,提供一种如上所述的CAR外泌体及其组合物在制备重症感染疾病或自身免疫性疾病治疗药物中的应用。
本发明的第五方面,提供一种制剂,所述的制剂为包含如上所述的CAR外泌体的组合物。
所述的制剂为包含CAR外泌体的组合物,在对包括人在内的动物给注射或以其他方式给药后,抗肿瘤效果明显。具体来讲,对肿瘤的预防和/或治疗有效,可以作为抗肿瘤药物使用。另外由于免疫细胞的性质,该种外泌体及外泌体组合物也可以对抗其他疾病,如重症感染疾病及自身免疫性疾病等。
本发明的第六方面,提供一种通过向患者给予治疗有效量的CAR外泌体来延长准备接受、正在接受或者已经接受癌症治疗(如利用化疗、放疗、靶向治疗及/或外科手术等)的癌症患者无复发生存期的方法。由于外泌体的组织穿透性,相比CAR免疫细胞的治疗方式,在实体瘤的治疗中应更具优势。
本发明中CAR外泌体及其组合物在对包括人在内的动物给药时,给药剂量因病人的年龄和体重,疾病特性和严重性,以及给药途径而异,可以参考动物实验的结果和种种情况,总给药量不能超过一定范围。
本发明优点在于:
本发明将CAR细胞(如CAR-T细胞等)以特异性的抗原活化后,产生的外泌体进行进一步的分析、分离、纯化、富集,最终得到携带CAR的免疫细胞外泌体。所述外泌体能用于各种疾病的治疗,如癌症,重型传染病等,且这种外泌体具克服CAR细胞治疗的免疫炎症风暴等不良反应能力,增强了CAR的组织浸透能力,还具有保存和运输方便的优势,为相关疾病的治疗提供了新的策略。
附图说明
图1.CAR外泌体抗原竞争性Elisa检测。
图2.CAR外泌体抗体竞争性Elisa检测。
图3.电子显微镜下重组EGFR蛋白活化的CAR-T来源的CAR外泌体形态。
图4.CAR-T来源的CAR外泌体抑制MDA-MB-231与HCC827细胞体外细胞生长。
图5.CAR-T来源的CAR外泌体抑制MDA-MB-231与HCC827细胞肿瘤生成曲线。
图6.电子显微镜下重组HER2蛋白活化的CAR-NK来源的CAR外泌体形态。
图7.CAR-NK来源的CAR外泌体及组合物抑制BT474与SK-BR-3细胞体外细胞生长。
图8.CAR-T来源的CAR外泌体对细胞的凋亡作用。
具体实施方式
下面结合实施例对本发明提供的具体实施方式作详细说明。
以下实施例、实验例对本发明进行进一步的说明,不应理解为对本发明的限制。实施例不包括对传统方法的详细描述,如那些用于构建载体和质粒的方法,将编码蛋白的基因插入到这样的载体和质粒的方法或将质粒引入宿主细胞的方法、或包装病毒的方法;利用病毒感染免疫细胞以获得目的蛋白表达的方法。这样的方法对于本领域中具有普通技术的人员是众所周知的,并且在许多出版物中都有所描述,包括Sambrook,J.,Fritsch,E.F.and Maniais,T.(1989)Molecular Cloning:A Laboratory Manual,2 ndedition,Cold spring Harbor Laboratory Press;Buchschacher,G.L.,Jr.,and Wong-Staal,F.(2000)Development of Lentiviral Vectors for Gene Therapy for Human Diseases.Blood 95,2499-2504;Yee,J.-K.,Miyanohara,A.,LaPorte,P.,Bouic,K.,Burns,J.C.,and Friedmann,T.(1994)A General Method for the Generation of High-Titer,Pantropic Retroviral Vectors:Highly Efficient Infection of Primary Hepatocytes.Proc.Natl.Acad.Sci.USA 91,9564-9568;Yee,J.K.(1999)in The Development of Human Gene Therapy(Friedmann,T.,ed),pp.21-45,Cold Spring Harbor Laboratory Press,Cold Spring Harbor,NY;Yee,J.K.,Moores,J.C.,Jolly,D.J.,Wolff,J.A.,Respess,J.G.,and Friedmann,T.(1987)Gene Expression from Transcriptionally Disabled Retroviral Vectors.Proc.Natl.Acad.Sci.USA 84,5197-5201;等。
实施例1.CAR-T细胞来源的CAR外泌体制备
1)全基因合成包含抗EGFR单链抗体的CAR序列(委托苏州金唯智生物科技有限公司合成),其中单链抗体序列来源于抗EGFR抗体cetuximab(Li et al.,2005,Structural basis for inhibition of the epidermal growth factor receptor by  cetuximab,Cancer Cell,7:301-311),CAR的具体结构包括:抗EGFR单链抗体scFv-CD8α铰链区及跨膜区域-4-1BB共激活结构域以及CD3ζ信号分子胞内段。具体序列与文献Johnson L A,et al.Science translational medicine,2015,7(275)报道的大体一致(除scFv)。为了方便检测,在scFv与铰链区之间插入Myc标签,标签的位置与文献Chu J,et al.CS1-specific chimeric antigen receptor(CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma.Leukemia,2014,28(4):917-927.相同。整个序列以同源重组的方式克隆进入含有CMV启动子的pLenti6.3/v5慢病毒载体(Invitrogen)中。
2)利用HEK293T细胞制备慢病毒。由于制备慢病毒是对于本领域中具有普通技术的人员是众所周知的,故不在此详述。简要的步骤包括取合适数量的HEK-293T细胞共转染构建好的慢病毒载体以及病毒包装质粒(Mission viral packaging plasmids,Sigma-Aldrich)。在转染72小时后收集病毒并浓缩(Lenti-X concentrator,Clontech)纯化病毒。
3)T细胞分离培养与CAR-T制备。通过密度梯度离心分离新鲜的外周血单核细胞(PBMC,peripheral blood mononuclear cell);利用偶联anti-CD3和anti-CD28抗体的paramagnetic beads(Dynabeads ClinExVivo CD3/CD28,Invitrogen,Camarillo,CA,USA)刺激并富集,磁珠与细胞比例为2-3:1;细胞稀释到浓度为5-8×l0 6/mL,在添加IL-2的培养基中共培育24小时;获得的T细胞以慢病毒多次感染。隔天计数细胞并更换培养基。当T细胞表现出静止态时进行下一步实验。所谓静止态指的是在细胞计数中增殖系数下降,细胞大小停止变化。此时收集转染前和转染后的细胞上清液提取外泌体作为对照使用。提取的方法后述。
4)T细胞特异性抗原激活。本步骤中使用两种方法进行CAR-T细胞特异性抗原激活,方法一是在T细胞培养基中加入磁珠偶联的EGFR胞外段重组蛋白。蛋白浓度为5ug/mL到1mg/mL之间分梯度进行实验。24小时候收集培养上清。上清中的重组蛋白通过磁场除去。方法二为CAR-T细胞与高表达EGFR的肿瘤细胞MDA-MB-231进行共培育。在培育前,MDA-MB-231细胞以100Gy的γ射线灭活。培育比例为2:1,4:1及8:1,24小时后收集培养上清。
5)外泌体制备。将3)和4)中描述的培养上清液按照如下步骤提取外泌体:将培养上清于500ml无菌离心瓶或50ml聚丙烯离心管中(购于Beckman 公司),于4℃、2000g离心10分钟,以去除死细胞和大的碎片。小心将上清液转移到新的无菌离心管中,于4℃、10000g离心30分钟,以去除细胞器及小颗粒。小心将上清液转移到无菌超速离心管中,于4℃、110000g(Beckman超速离心机)超速离心70分钟,小心弃去上清,再加注射用生理盐水清洗一次,于4℃、110,000g超速离心70分钟,得到的沉淀即为外泌体。根据最初收集的培养基的体积不同,酌情加入PBS重悬。
6)携带CAR外泌体制备
将获得的外泌体按照以下步骤纯化富集携带CAR的外泌体:向含有外泌体的生理盐水溶液中加入有EGFR胞外段重组蛋白包被的磁珠,4℃孵育30min,磁珠通过特异性抗原抗体相互作用与带有相应svFv的CAR外泌体结合;将试管置于磁场中,与磁珠连接的外泌体被磁场吸附;磁珠被吸附后,吸去上清,将试管移出磁场;用PBS缓冲液悬浮重悬,加入分选柱中,收集先行流出的未结合组分,并用缓冲液冲洗分选柱,洗下来的为不携带有CAR的外泌体囊泡。将分选柱移出磁场,用缓冲液快速将分选柱上滞留的携带有CAR的外泌体洗脱下来,洗脱下来的即为目的外泌体。用Bradford试剂盒(购于Thermo公司)检测总蛋白浓度后,可以在-80摄氏度分装长期保存。
值得说明的是,利用未经特异性抗原刺激的CAR-T细胞分泌的外泌体进行纯化富集无法获得可用于后续实现的CAR外泌体。即未经特异性抗原刺激的CAR-T细胞分泌的外泌体中的携带CAR的外泌体含量是极少的。
7)携带CAR外泌体检测。
由于ELISA实验为本领域的常见实验,为一般的技术人员所熟知,下列实施例中未注明具体条件的实验方法,可采用本领域中的常规方法,例如参考《分子克隆实验指南》(第三版,纽约,冷泉港实验室出版社,New York:Cold Spring Harbor Laboratory Press,1989)或按照供应商所建议试剂盒步骤操作。按照如下步骤检测外泌体的CAR表达:按照一定的稀释比例的CAR外泌体加入到包裹有EGFR抗原的封闭完成的96孔板中,37℃孵育1h。洗涤后按照竞争的方式加入EGFR重组蛋白,共孵育。TBST洗涤3次,后加入HRP标记的Anti-Myc抗体。37℃孵育1h。TBST洗涤后,加入显色底物。酶标仪检测后计算分析,结果如图1。与Cetuximab的竞争ELISA如下:按照一定的稀释比例的CAR外泌体和biotin标记的Cetuximab加入到包裹有EGFR抗原的封闭完 成的96孔板中,37℃孵育1h。TBST洗涤3次,后加入HRP标记的亲和素(Thermo)。37℃孵育1h。TBST洗涤后,加入显色底物。酶标仪检测后计算分析,结果如图2。
透射电镜观察获得的CAR外泌体的形态:将外泌体充分重悬,吸取10μ1滴到载样铜网上,室温静置5分钟,用滤纸小心吸掉多余液体。滴加醋酸双氧铀负染2分钟,用滤纸吸掉多余液体,在白炽灯下烘干;透射电镜80-120kv成像并拍照。结果如图所示,可见直径为30-150nm左右的圆形囊泡状结构,结果如图3。
该实施例结果说明,成功获取免疫细胞来源的携带有CAR的外泌体,它们都表达CAR蛋白,能结合特异性的抗原,平均直径在80nm左右,透射电镜下观察到的形态都符合外泌体的特征。
实施例2:CAR外泌体抑制EGFR阳性的MDA-MB-231及HCC827细胞活力实验
取生长状态良好的MDA-MB-231及HCC827细胞(ATCC),调整细胞浓度为5×10 3/ml,接种于96孔细胞培养板,200μl/孔,于37℃、5%CO 2孵箱中培养24h后,在培养液中加入终浓度为5nmol的EGF和不同浓度梯度的外泌体,另用cetuximab抗体药物做对照(Cetuximab购自默克公司),4天后,细胞活力用CellTiter-Glo Luminescent Cell Viability Assay试剂盒(Promega,Madison,WI)检测。实验结果如图3所示。实验结果表明,CAR外泌体可以显著抑制的MDA-MB-231和HCC827细胞活力(P<0.01,Tukey检验),尤其是对cetuximab天然耐药的MDA-MB-231细胞。(图4)。
实施例3:CAR外泌体体内抑制肿瘤生长实验
为检测CAR外泌体体内抑瘤活性,首先用HCC827和MDA-231细胞,接种于到BALB/c裸鼠(中科院实验动物中心)右胁侧皮下,成瘤后尾静脉注射3500mg/kg的CAR外泌体及抗体药物cetuximab(10mg/kg),每周注射1次,持续至小鼠肿瘤过大处死。每天测量肿瘤的长宽,计算肿瘤体积。
肿瘤生长曲线如图5所示。结果表明激活型CAR外泌体治疗组肿瘤生长速度显著小于cetuximab治疗组(40天后,P<0.01,Bonferroni检验)。
实施例4:靶向HER2的CAR-NK细胞来源的CAR外泌体制备
1)全基因合成包含抗HER2单链抗体的CAR序列(委托苏州金唯智生物 科技有限公司合成),其中单链抗体序列来源于抗HER2抗体trastuzumab(Cho H S,et al.Structure of the extracellular region of HER2alone and in complex with the Herceptin Fab.Nature,2003,421(6924):756-760.),CAR的具体结构包括:抗HER2单链抗体scFv-CD28铰链区及跨膜区域CD28以及CD3ζ信号分子胞内段。具体序列同文献Chu J,et al.CS1-specific chimeric antigen receptor(CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma.Leukemia,2014,28(4):917-927.(除ScFv段)。整个序列以同源重组的方式克隆进入含有CMV启动子的PCDH慢病毒载体(System Biosciences)中。
2)利用HEK293T细胞制备慢病毒。由于制备慢病毒是对于本领域中具有普通技术的人员是众所周知的,故不在此详述。简要的步骤包括取合适数量的HEK-293T细胞共转染构建好的慢病毒载体以及病毒包装质粒pCMV-VSVG和pCMV-dr9。在转染72小时后收集病毒并浓缩(Lenti-X concentrator,Clontech)纯化病毒。
3)CAR-NK制备。
将NK-92细胞稀释到1×10 6/mL并在添加IL-2的培养基中培养过夜,然后以慢病毒反复连续感染。在第三次感染后,将细胞培养至含有20%FBS的1640培养基中。该培养基添加150单位/ml的IL-2。经过两次流式细胞术分选(BD Biosciences,San Jose,CA,USA)绿色荧光蛋白表达的细胞。该绿色荧光蛋白为PCDH载体基因所编码。另外在实验中收集感染前后的细胞上清液提取外泌体作为对照使用。提取的方法后述。
4)NK细胞特异性抗原激活。本步骤中使用两种种方法进行NK细胞特异性抗原激活,方法一是在NK细胞培养基中加入磁珠偶联的HER2胞外段重组蛋白。蛋白浓度为5ug/mL到1mg/mL之间分梯度进行实验。12-24小时候收集培养上清。方法二为NK细胞与高表达HER2的灭活后BT474细胞进行共培育。培育比例为2:1,4:1及8:1,12-24小时后收集培养上清。
5)外泌体制备。将3)和4)中描述的培养上清液按照如下步骤提取外泌体:将培养上清于500ml无菌离心瓶或50ml聚丙烯离心管中(购于Beckman公司),于4℃、2000g离心10分钟,以去除死细胞和大的碎片。小心将上清液转移到新的无菌离心管中,于4℃、10000g离心30分钟,以去除细胞器及小颗粒。小心将上清液转移到无菌超速离心管中,于4℃、110000g(Beckman 超速离心机)超速离心70分钟,小心弃去上清,再加注射用生理盐水清洗一次,于4℃、110,000g超速离心70分钟,得到的沉淀即为外泌体。根据最初收集的培养基的体积不同,酌情加入PBS重悬。
6)携带CAR外泌体制备
将获得的外泌体按照以下步骤纯化富集携带CAR的外泌体:首先向含有外泌体的生理盐水溶液中加入包被有蛋白L的磁珠,4℃孵育60min,磁珠通过蛋白L与免疫球蛋白轻链的特异性结合力与CAR外泌体或含有免疫球蛋白的外泌体结合;将试管置于磁场中,与磁珠连接的外泌体被磁场吸附;磁珠被吸附后,吸去上清,将试管移出磁场;用PBS缓冲液悬浮重悬,加入分选柱中,收集先行流出的未结合组分,并用缓冲液冲洗分选柱,洗下来的为不携带有免疫球蛋白的外泌体囊泡。将分选柱移出磁场,用缓冲液快速将分选柱上滞留的外泌体洗脱下来后立即与重组HER2蛋白包被的磁珠,4℃孵育30min,磁珠通过特异性抗原抗体相互作用与带有相应svFv的CAR外泌体结合;将试管置于磁场中,与磁珠连接的外泌体被磁场吸附;磁珠被吸附后,吸去上清,将试管移出磁场;用PBS缓冲液悬浮重悬,加入分选柱中,收集先行流出的未结合组分,并用缓冲液冲洗分选柱,洗下来的为不携带有CAR的外泌体囊泡。将分选柱移出磁场,用缓冲液快速将分选柱上滞留的携带有CAR的外泌体洗脱下来并平衡至生理pH,洗脱下来的即为目的外泌体。用Bradford试剂盒(购于Thermo公司)检测总蛋白浓度后,可以在-80摄氏度分装长期保存。
7)携带CAR外泌体检测。
透射电镜观察获得的CAR外泌体的形态:将外泌体充分重悬,吸取10ul滴到载样铜网上,室温静置5分钟,用滤纸小心吸掉多余液体。滴加醋酸双氧铀负染2分钟,用滤纸吸掉多余液体,在白炽灯下烘干;透射电镜80-120kv成像并拍照。结果如图所示,可见直径为30-150nm左右的圆形囊泡状结构,结果如图6。
实施例5:NK来源的CAR外泌体抑制乳腺癌细胞活力实验
取生长状态良好的HER2高表达乳腺癌细胞系BT474及HER2低表达MCF-7细胞(ATCC),调整细胞浓度为4×10 3/ml,接种于96孔细胞培养板,200μl/孔,于37℃、5%CO 2孵箱中培养24h后,在培养液中加入不同浓度梯度的外泌体,另用trastuzumab抗体药物做对照,4天后,细胞活力用 CellTiter-Glo Luminescent Cell Viability Assay试剂盒(Promega,Madison,WI)检测。实验结果如图7所示。实验结果表明,NK细胞来源CAR外泌体可以显著抑制BT474细胞活力(P<0.01,Tukey检验),但对HER2低表达的MCF-7细胞抑制作用较弱。(图7)。
实施例6:负载阿霉素的NK来源的CAR外泌体制备及其抗肿瘤效果
将实施例4和5中获取的CAR外泌体与等质量的阿霉素化疗药物混合。通过电转的方法制备化合物负载CAR外泌体。电击条件为电压420V,电容150μF,于4mm电转杯进行电转。随后用倒置离心超滤膜过滤,去除未转染进外泌体内的游离化合物。
也可以通过脂质体转染法将化合物导入外泌体实现对外泌体的负载。取生长状态良好的HER2高表达乳腺癌细胞系BT474及HER2低表达MCF-7细胞(ATCC),调整细胞浓度为4×10 3/ml,接种于96孔细胞培养板,200μl/孔,于37℃、5%CO 2孵箱中培养24h后,在培养液中加入不同浓度梯度的外泌体,,4天后,细胞活力用CellTiter-Glo Luminescent Cell Viability Assay试剂盒(Promega,Madison,WI)检测。实验结果如图7所示。实验结果表明,负载阿霉素的CAR外泌体可以更加显著抑制肿瘤细胞活力(P<0.01,Tukey检验)。
实施例7:靶向CD20的CAR-T来源的CAR外泌体对淋巴瘤细胞凋亡作用。
1)全基因合成包含抗CD20单链抗体的CAR序列(委托苏州金唯智生物科技有限公司成),其中单链抗体序列来源于抗CD抗体Rituximab(Du J,et al.Structural basis for recognition of CD20by therapeutic antibody Rituximab.Journal of Biological Chemistry,2007,282(20):15073-15080.)。CAR的具体结构包括:抗CD20单链抗体scFv-铰链区及CD28跨膜区域CD28、4-1BB以及CD3ζ信号分子胞内段。具体序列同文献Chu J,et al.CS1-specific chimeric antigen receptor(CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma.Leukemia,2014,28(4):917-927.(除ScFv、4-1BB)。整个序列以同源重组的方式克隆进入含有CMV启动子的PCDH慢病毒载体(System Biosciences)中。
利用HEK293T细胞制备慢病毒的方法、制备CAR-T细胞的方法、CAR-T细胞特异性抗原激活刺激的方法、外泌体制备及携带CAR外泌体的纯化方法同上述实施例,故在此不再赘述。其中,CAR-T细胞的特异性抗原活化剂为表 达CD20的灭活Raji细胞。
取生长状态良好的Burkitt淋巴瘤细胞系Raji(ATCC),调整细胞浓度为1×10 5/孔。于37℃、5%CO 2孵箱中培养24小时后后,在培养液中加入不同浓度梯度的外泌体,另用Rituximab抗体药物做对照,培养16小时候对细胞进行漂洗,然后用annexin V-FITC(BD Biosciences)染色后行流式细胞术计算凋亡细胞比率。实验结果如图8所示。实验结果表明,CAR-T细胞来源CAR外泌体可以明显诱导Raji细胞及Daudi细胞凋亡(P<0.01,Tukey检验)。(图8)。
以上已对本发明创造的较佳实施例进行了具体说明,但本发明创造并不限于所述实施例,熟悉本领域的技术人员在不违背本发明创造精神的前提下还可做出种种的等同的变型或替换,这些等同的变型或替换均包含在本申请权利要求所限定的范围内。

Claims (10)

  1. 一种携带CAR的免疫细胞外泌体的制备方法,其特征在于,包括以下步骤:
    A)制备CAR免疫细胞:
    按照通用的生物工程手段制备CAR免疫细胞;
    B)对CAR免疫细胞进行抗原特异性活化:
    采用的活化剂是特异性靶点的可溶性重组蛋白、表达特异性靶点的工程细胞、或直接使用表达特异性靶点肿瘤细胞;所述的特异性靶点指的是CAR免疫细胞中的scFv针对的抗原靶点,即CAR免疫细胞靶向的特异性靶点;活化的方法为在体外的培养体系中加入抗原蛋白或固化的抗原蛋白、直接共培育CAR免疫细胞及和表达特异性靶点的灭活后工程细胞、直接共培育CAR免疫细胞和表达特异性靶点的灭活后肿瘤细胞;
    C)收集制备CAR免疫细胞外泌体:
    采集培养上清,按照通用的外泌体分离方法进行收集制备;
    D)CAR外泌体的纯化和富集:
    向步骤C中制得的外泌体重悬液中加入特异性抗原包被的磁性体,即CAR捕获磁体,所述磁性体上含有与CAR蛋白特异性结合的重组靶点蛋白抗原;孵育后将重悬液置于磁场中,除去上清液后加入缓冲液,加入分选柱,利用缓冲液洗脱分选柱上滞留的外泌体;将上清液加入分选柱后,先流出的是无抗原结合能力的外泌体,然后采用缓冲液洗脱分选柱,流出的即为具有抗原结合能力、携带CAR的免疫细胞外泌体;根据最初收集的培养基的体积不同,酌情加入PBS重悬,用Bradford试剂盒检测总蛋白浓度,-80℃分装保存。
  2. 根据权利要求1所述的携带CAR的免疫细胞外泌体的制备方法,其特征在于,所述的步骤A的免疫细胞是T细胞或NK细胞;所述的免疫细胞的来源是患者自身,或健康志愿者。
  3. 根据权利要求1所述的携带CAR的免疫细胞外泌体的制备方法,其特征在于,所述的步骤A的免疫细胞为健康志愿者来源的T细胞,CAR免疫细胞的制备方法按照以下步骤实施:
    (a)获得细胞样本并分离和激活,所述样本为T细胞或T细胞的祖细胞;
    (b)构建scFv-CD8hinge and TM-4-1BB-CD3、scFv-hinge-TM-CD28-CD3、scFv-hinge-CD28-4-1BB-CD3的病毒载体;
    (c)制作重组质粒并包装病毒;
    (d)病毒感染T细胞、NK细胞;
    (e)体外培养扩增CAR-T、CAR-NK细胞群体。
  4. 根据权利要求1所述的携带CAR的免疫细胞外泌体的制备方法,其特征在于,所述的步骤B中CAR免疫细胞中的scFv针对的抗原靶点是EGFR,HER2,CD20。
  5. 根据权利要求1所述的携带CAR的免疫细胞外泌体的制备方法,其特征在于,所述的步骤B采用的活化剂为表皮生长因子EGFR胞外段重组蛋白、磁珠交联的EGFR胞外段重组蛋白,表达EGFR的CHO细胞、表达EGFR的MDA-MB-231细胞;磁珠交联的HER2胞外段重组蛋白、表达HER2的BT474细胞、表达CD20的Raji细胞。
  6. 根据权利要求1所述的携带CAR的免疫细胞外泌体的制备方法,其特征在于,所述的步骤C收集制备CAR免疫细胞外泌体的方法为:将收集的培养上清于4℃、2000g离心10分钟,以去除死细胞和大的碎片;小心将上清液转移到新的无菌离心管中,于4℃、10000g离心30分钟,以去除细胞器及小颗粒;小心将上清液转移到无菌超速离心管中,于4℃,110000g超速离心70分钟,小心弃去上清,再加PBS清洗一次,于4℃,110000g超速离心70分钟,得到的沉淀即为外泌体。
  7. 一种采用如权利要求1-6任一所述的制备方法制备得到的携带CAR的免疫细胞外泌体,所述的外泌体携带CAR蛋白,平均直径在30-150nm左右。
  8. 一种如权利要求7所述的携带CAR的免疫细胞外泌体及其组合物在制备抗肿瘤药物中的应用。
  9. 一种如权利要求7所述的携带CAR的免疫细胞外泌体及其组合物在制备重症感染疾病或自身免疫性疾病治疗药物中的应用。
  10. 一种制剂,所述的制剂为包含如权利要求7所述的携带CAR的免疫细胞外泌体的组合物。
PCT/CN2018/123298 2017-12-26 2018-12-25 携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用 WO2019128952A1 (zh)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/043,670 US20210030801A1 (en) 2017-12-26 2018-12-25 Method for preparing chimeric antigen receptor (car)-carrying exosomes derived from immune cells, and use of car-carrying exosomes
EP18897524.7A EP3747995B1 (en) 2017-12-26 2018-12-25 METHOD FOR PREPARING CHIMERIC ANTIGEN RECEPTOR &#xA;(CAR)-CARRYING EXOSOMES DERIVED FROM IMMUNE CELLS, AND USE OF SAID CAR- CARRYING EXOSOMES

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201711432948.4 2017-12-26
CN201711432948.4A CN108315305B (zh) 2017-12-26 2017-12-26 携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用

Publications (1)

Publication Number Publication Date
WO2019128952A1 true WO2019128952A1 (zh) 2019-07-04

Family

ID=62893576

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/123298 WO2019128952A1 (zh) 2017-12-26 2018-12-25 携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用

Country Status (4)

Country Link
US (1) US20210030801A1 (zh)
EP (1) EP3747995B1 (zh)
CN (1) CN108315305B (zh)
WO (1) WO2019128952A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020212985A1 (en) 2019-04-18 2020-10-22 Yeda Research And Development Co. Ltd. Extracellular vesicles derived from activated car-t cells
WO2021184673A1 (en) * 2020-03-17 2021-09-23 Cellular Biomedicine Group Hk Limited Combined chimeric antigen receptor targeting cd19 and cd20 and application thereof
CN114259568A (zh) * 2021-10-12 2022-04-01 厦门大学 外泌体在制备阿霉素和氯尼达明联合递送制剂中的应用

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108315305B (zh) * 2017-12-26 2020-11-06 沣潮医药科技(上海)有限公司 携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用
EP3912629A4 (en) * 2018-12-17 2022-10-12 Cure Genetics Co., Ltd METHOD OF GENE DELIVERY INTO CELLS
CN109666074B (zh) * 2018-12-29 2020-04-24 广州百暨基因科技有限公司 一种趋化因子受体cxcr5的用途
WO2020180744A1 (en) * 2019-03-01 2020-09-10 The Regents Of The University Of California Natural killer cell induced cellular vesicles for cancer therapy
CN110358737B (zh) * 2019-07-24 2021-06-29 天津市中西医结合医院(天津市南开医院) 一种利用外泌体制备嵌合抗原受体t淋巴细胞的方法
US20220409741A1 (en) * 2019-09-02 2022-12-29 Kyungpook National University Industry-Academic Cooperation Foundation Composition for preventing or treating cancer, containing il-2 surface expression-extracellular vesicles as active ingredient
CN110964750A (zh) * 2019-11-13 2020-04-07 沣潮医药科技(上海)有限公司 携带基因元件组合的嵌合抗原受体细胞文库、制备和筛选方法及用途
CN111135307B (zh) * 2020-02-05 2023-05-30 中山大学附属第八医院(深圳福田) 一种基于car-t细胞的药物传递系统的制备方法及应用
CN112574953A (zh) * 2020-12-11 2021-03-30 南通大学 一种间皮素嵌合抗原受体外泌体、及其制备方法和应用
EP4115892A1 (en) * 2021-07-07 2023-01-11 Fundación Pública Andaluza Progreso Y Salud In vitro method for improving the production of exosomes from car-t cells
CN116286948A (zh) * 2023-02-21 2023-06-23 河南科技大学 一种可活化t细胞的盐藻外泌体组合物的制备方法及应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017011728A1 (en) * 2015-07-15 2017-01-19 Washington University ANTIBODIES TO TUMOR ASSOCIATED COMPLEX N-GLYCANS WITH TERMINAL GlcNAcBeta RESIDUES AND METHODS OF USE THEREOF
CN108315305A (zh) * 2017-12-26 2018-07-24 沣潮医药科技(上海)有限公司 携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103740645A (zh) * 2013-12-06 2014-04-23 山西医科大学 神经干细胞来源Exosomes制备及在神经系统疾病中的应用
CN105384824A (zh) * 2014-08-26 2016-03-09 中国人民解放军总医院 嵌合抗原受体及其基因和重组表达载体、工程化her2靶向性的nkt细胞及其应用
CN105861531B (zh) * 2016-04-21 2021-04-06 汪治宇 一种嵌合抗原受体t细胞及其制备方法

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017011728A1 (en) * 2015-07-15 2017-01-19 Washington University ANTIBODIES TO TUMOR ASSOCIATED COMPLEX N-GLYCANS WITH TERMINAL GlcNAcBeta RESIDUES AND METHODS OF USE THEREOF
CN108315305A (zh) * 2017-12-26 2018-07-24 沣潮医药科技(上海)有限公司 携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用

Non-Patent Citations (30)

* Cited by examiner, † Cited by third party
Title
AHMED N ET AL.: "Human Epidermal Growth Factor Receptor 2 (HER2)-Specific Chimeric Antigen Receptor-Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma", JOURNAL OF CLINICAL ONCOLOGY, vol. 33, no. 15, 2015, pages 1688 - 1696, XP055448271, DOI: 10.1200/JCO.2014.58.0225
BRENTJENS R ET AL.: "Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial", MOLECULAR THERAPY, vol. 18, no. 4, 2010, pages 666 - 668, XP055143817, DOI: 10.1038/mt.2010.31
BUCHSCHACHER, G. L., JR.WONG-STAAL, F.: "Development of Lentiviral Vectors for Gene Therapy for Human Diseases", BLOOD, vol. 95, 2000, pages 2499 - 2504
CARPENITO C ET AL.: "Control of Large, Established Tumor Xenografts With Genetically Retargeted Human T Cells Containing CD28 and CD137 Domains", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 106, no. 9, 2009, pages 3360 - 3365
CARUSO H G ET AL.: "Tuning sensitivity of CAR to EGFR density limits recognition of normal tissue while maintaining potent antitumor activity[J", CANCER RESEARCH, vol. 75, no. 17, 2015, pages 3505 - 3518, XP055357260, DOI: 10.1158/0008-5472.CAN-15-0139
CHO H S ET AL.: "Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab", NATURE, vol. 421, no. 6924, 2003, pages 756 - 760, XP009113017, DOI: 10.1038/nature01392
CHU J ET AL.: "CS 1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma", LEUKEMIA, vol. 28, no. 4, 2014, pages 917 - 927, XP055133640, DOI: 10.1038/leu.2013.279
CHU J ET AL.: "CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma", LEUKEMIA, vol. 28, no. 4, 2014, pages 917 - 927, XP055133640, DOI: 10.1038/leu.2013.279
CHU J ET AL.: "CSl-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma", LEUKEMIA, vol. 28, no. 4, 2014, pages 917 - 927, XP055133640, DOI: 10.1038/leu.2013.279
COUMANS F A W ET AL.: "Methodological Guidelines to Study Extracellular Vesicles", CIRCULATION RESEARCH, vol. 120, no. 10, 2017, pages 1632 - 1648, XP055520220, DOI: 10.1161/CIRCRESAHA.117.309417
DU J ET AL.: "Structural basis for recognition of CD20 by therapeutic antibody Rituximab", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 282, no. 20, 2007, pages 15073 - 15080, XP055094566, DOI: 10.1074/jbc.M701654200
JOHNSON L A ET AL., SCIENCE TRANSLATIONAL MEDICINE, vol. 7, no. 275, 2015
JOHNSON L A ET AL.: "Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma", SCIENCE TRANSLATIONAL MEDICINE, vol. 7, no. 275, 2015, pages 275 - 22,275-22, XP055362795, DOI: 10.1126/scitranslmed.aaa4963
KOOIJMANS, S.A.A. ET AL.: "Display of GPI-Anchored Anti-EGFR Nanobodies on Extracellular Vesicles Promotes Tumour Cell Targeting", JOURNAL OF EXTRACELLULAR VESICLES, 16 January 2016 (2016-01-16), pages 31053, XP055416987, ISSN: 2001-3078 *
LI ET AL.: "Structural basis for inhibition of the epidermal growth factor receptor by cetuximab", CANCER CELL, vol. 7, 2005, pages 301 - 311, XP002508255, DOI: 10.1016/J.CCR.2005.03.003
LI L ET AL.: "Human bile contains MicroRNA-laden extracellular vesicles that can be used for cholangiocarcinoma diagnosis", HEPATOLOGY, vol. 60, no. 3, 2014, pages 896 - 907, XP055476444, DOI: 10.1002/hep.27050
LI LPIONTEK KISHIDA M ET AL.: "Extracellular vesicles carry microRNA-195 to intrahepatic cholangiocarcinoma and improve survival in a rat model", HEPATOLOGY, vol. 65, no. 2, 2017, pages 501 - 514, XP055476283, DOI: 10.1002/hep.28735
LI N ET AL.: "Therapeutically targeting glypican-2 via single-domain antibody-based chimeric antigen receptors and immunotoxins in neuroblastoma", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 114, no. 32, 2017, pages E6623 - E6631, XP055404734, DOI: 10.1073/pnas.1706055114
PARK S ET AL.: "Micromolar affinity CAR T cells to ICAM-1 achieves rapid tumor elimination while avoiding systemic toxicity", SCIENTIFIC REPORTS, vol. 7, no. 1, 2017, pages 14366, XP055656078, DOI: 10.1038/s41598-017-14749-3
SAMBROOK, J.FRITSCH, E. F.MANIAIS, T.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SAVOLDO B ET AL.: "CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 121, no. 5, 2011, pages 1822, XP055204919, DOI: 10.1172/JCI46110
See also references of EP3747995A4
TANG X J ET AL.: "Therapeutic potential of CAR-T cell-derived exosomes: a cell-free modality for targeted cancer therapy", ONCOTARGET, vol. 6, no. 42, 2015, pages 44179, XP055585353, DOI: 10.18632/oncotarget.6175
TANG, X.J. ET AL.: "Therapeutic Potential of CAR-T Cell -Derived Exosomes: a Cell -Free Modality for Targeted Cancer Therapy", ONCOTARGET, vol. 6, no. 42, 29 December 2015 (2015-12-29), pages 44179 - 44190, XP055585353, ISSN: 1949-2553 *
THERY C ET AL.: "Isolation and characterization of exosomes from cell culture supernatants and biological fluids", CURRENT PROTOCOLS IN CELL BIOLOGY, vol. 2006
TURTLE C J ET AL.: "CD 19 CAR-T cells of defined CD4+: CD8+ composition in adult B cell ALL patients", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 126, no. 6, 2016, pages 2123, XP055402477, DOI: 10.1172/JCI85309
YEE, J. K.: "The Development of Human Gene Therapy", 1999, COLD SPRING HARBOR LABORATORY PRESS, pages: 21 - 45
YEE, J. K.MOORES, J. C.JOLLY, D. J.WOLFF, J. A.RESPESS, J. G.FRIEDMANN, T.: "Gene Expression from Transcriptionally Disabled Retroviral Vectors", PROC. NATL. ACAD. SCI. USA, vol. 84, 1987, pages 5197 - 5201, XP008108549, DOI: 10.1073/pnas.84.15.5197
YEE, J.-K.MIYANOHARA, A.LAPORTE, P.BOUIC, K.BURNS, J. C.FRIEDMANN, T.: "A General Method for the Generation of High-Titer, Pantropic Retroviral Vectors: Highly Efficient Infection of Primary Hepatocytes", PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 9564 - 9568, XP000674728, DOI: 10.1073/pnas.91.20.9564
ZHANG TBARBER ASENTMAN C L.: "Chimeric NKG2D-Modified T Cells Inhibit Systemic T-Cell Lymphoma Growth in a Manner Involving Multiple Cytokines and Cytotoxic Pathways[J", CANCER RESEARCH, vol. 67, no. 22, 2007, pages 11029 - 11036

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020212985A1 (en) 2019-04-18 2020-10-22 Yeda Research And Development Co. Ltd. Extracellular vesicles derived from activated car-t cells
WO2021184673A1 (en) * 2020-03-17 2021-09-23 Cellular Biomedicine Group Hk Limited Combined chimeric antigen receptor targeting cd19 and cd20 and application thereof
US11207349B2 (en) 2020-03-17 2021-12-28 Cellular Biomedicine Group Hk Limited Combined chimeric antigen receptor targeting CD19 and CD20 and application thereof
CN114259568A (zh) * 2021-10-12 2022-04-01 厦门大学 外泌体在制备阿霉素和氯尼达明联合递送制剂中的应用

Also Published As

Publication number Publication date
US20210030801A1 (en) 2021-02-04
CN108315305B (zh) 2020-11-06
EP3747995A4 (en) 2021-06-02
EP3747995A1 (en) 2020-12-09
CN108315305A (zh) 2018-07-24
EP3747995B1 (en) 2022-11-09

Similar Documents

Publication Publication Date Title
WO2019128952A1 (zh) 携带嵌合抗原受体的免疫细胞外泌体的制备方法及其应用
Suarez et al. Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model
Schönfeld et al. Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor
CN109400713B (zh) 新型嵌合抗原受体修饰的t细胞治疗癌症的用途
Katsarou et al. Combining a CAR and a chimeric costimulatory receptor enhances T cell sensitivity to low antigen density and promotes persistence
WO2017032293A1 (zh) 抗间皮素全人抗体以及靶向间皮素的免疫效应细胞
CN108409840B (zh) 抗cd123单链抗体及其组合的嵌合抗原受体和应用
An et al. Construction of a new anti-CD19 chimeric antigen receptor and the anti-leukemia function study of the transduced T cells
Muliaditan et al. Synergistic T cell signaling by 41BB and CD28 is optimally achieved by membrane proximal positioning within parallel chimeric antigen receptors
CN112426526B (zh) 一种nk细胞的制备方法及其在治疗癌症中的应用
KR20190003456A (ko) 면역치료의 항-종양 활성을 높이기 위한 중간엽 줄기세포
KR20200015717A (ko) 암 치료를 위한 인간 종양 반응성 t 세포의 확인을 위한 cd39 및 cd103의 활용
CN109021114B (zh) 联合两种单链抗体的双特异性嵌合抗原受体及表达载体
WO2020248486A1 (zh) 以Tcm为主要效应成分的CAR-T制备方法及其应用
KR20210078518A (ko) 조직 거주 기억-유사 t 세포의 생산 방법 및 이의 용도
US11014974B2 (en) Non-antibody binding proteins binding to PD-1 receptors and uses thereof
CN109232740B (zh) 一种抗pd-l1抗体及其在抗肿瘤治疗中的应用
Qiao et al. Enhancement of CAR‐T cell activity against cholangiocarcinoma by simultaneous knockdown of six inhibitory membrane proteins
Wu et al. A CH2CH3 hinge region enhances the cytotoxicity of anti-CD5 CAR-T cells targeting T cell acute lymphoblastic leukemia
WO2022171195A1 (zh) 抗cd87抗体与抗pd1抗体联合治疗胃癌
Hussein et al. TCR T cells overexpressing c-Jun have better functionality with improved tumor infiltration and persistence in hepatocellular carcinoma
KR20230030587A (ko) Nk 세포와 항체의 결합을 안정화시키는 방법, 및 그 이용
Sun et al. A dendritic/tumor fusion cell vaccine enhances efficacy of nanobody-based CAR-T cells against solid tumor
Potez et al. Use of phage display biopanning as a tool to design CAR-T cells against glioma stem cells
CN116284448B (zh) 一种超抗原参与的三功能t细胞衔接器及其应用

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: 18897524

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018897524

Country of ref document: EP

Effective date: 20200727