CN114931632A - Cancer vaccine based on antigen presenting cell membrane component and preparation method and application thereof - Google Patents
Cancer vaccine based on antigen presenting cell membrane component and preparation method and application thereof Download PDFInfo
- Publication number
- CN114931632A CN114931632A CN202210630310.6A CN202210630310A CN114931632A CN 114931632 A CN114931632 A CN 114931632A CN 202210630310 A CN202210630310 A CN 202210630310A CN 114931632 A CN114931632 A CN 114931632A
- Authority
- CN
- China
- Prior art keywords
- antigen
- cells
- cancer
- interleukin
- presenting
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/0005—Vertebrate antigens
- A61K39/0011—Cancer antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/16—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
- A61K47/18—Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
- A61K47/183—Amino acids, e.g. glycine, EDTA or aspartame
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/26—Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/30—Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
- A61K47/32—Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/30—Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
- A61K47/42—Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/69—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
- A61K47/6901—Conjugates being cells, cell fragments, viruses, ghosts, red blood cells or viral vectors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/14—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
- A61K9/19—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/50—Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
- A61K9/5005—Wall or coating material
- A61K9/5063—Compounds of unknown constitution, e.g. material from plants or animals
- A61K9/5068—Cell membranes or bacterial membranes enclosing drugs
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/515—Animal cells
- A61K2039/5154—Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/06—Anti-neoplasic drugs, anti-retroviral drugs, e.g. azacytidine, cyclophosphamide
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Epidemiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Zoology (AREA)
- Organic Chemistry (AREA)
- Microbiology (AREA)
- Biomedical Technology (AREA)
- Cell Biology (AREA)
- Biochemistry (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Molecular Biology (AREA)
- Hematology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Oil, Petroleum & Natural Gas (AREA)
- Virology (AREA)
- Inorganic Chemistry (AREA)
- Biotechnology (AREA)
- Mycology (AREA)
- Oncology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biophysics (AREA)
- Botany (AREA)
- General Engineering & Computer Science (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicinal Preparation (AREA)
Abstract
本发明涉及一种基于抗原提呈细胞膜组分的癌症疫苗及其制备方法和应用,癌症疫苗包括以下其中之一:(1)由激活的抗原提呈细胞细胞膜组分制备得到的纳米囊泡;(2)内部和/或外部负载癌细胞全细胞组分,且表面负载激活的抗原提呈细胞细胞膜组分的第二递送粒子。本发明在实现源于树突状细胞的疫苗负载广谱多样的癌细胞抗原的同时克服了树突状细胞活性保持困难、无法冻干长期储存等活细胞疫苗的难题,能够制备负载广谱癌细胞抗原表位的癌症疫苗,用于癌症的预防和治疗。
The present invention relates to a cancer vaccine based on antigen-presenting cell membrane components and a preparation method and application thereof. The cancer vaccine includes one of the following: (1) nanovesicles prepared from activated antigen-presenting cell membrane components; (2) A second delivery particle that internally and/or externally loads the whole cell component of the cancer cell, and the surface loads the activated antigen-presenting cell membrane component. The invention realizes that the vaccine derived from dendritic cells can be loaded with broad-spectrum and diverse cancer cell antigens, and at the same time overcomes the difficulties of maintaining the activity of dendritic cells and the inability to freeze-dry long-term storage of live cell vaccines, and can prepare a broad-spectrum cancer-bearing vaccine. Cancer vaccines based on cellular epitopes for the prevention and treatment of cancer.
Description
技术领域technical field
本发明涉及免疫治疗领域,尤其涉及一种基于抗原提呈细胞膜组分的癌症疫苗及其制备方法和应用。The invention relates to the field of immunotherapy, in particular to a cancer vaccine based on antigen-presenting cell membrane components and a preparation method and application thereof.
背景技术Background technique
癌症免疫治疗是癌症最主要的治疗方法之一,其中癌症疫苗是癌症免疫治疗的重要方法之一。癌症疫苗通常的作用机理是:癌症疫苗负载的抗原被抗原提呈细胞吞噬降解,将降解成小短肽的抗原表位与主要组织相容性复合物(MHC)分子结合后提呈到抗原提呈细胞表面以激活癌细胞特异性T细胞。癌症疫苗通过抗原提呈细胞将Naive T细胞激活成癌细胞特异性T细胞的过程属于T细胞的初次激活,需要依靠树突状细胞(DC)才能完成,且在淋巴结中完成。一旦T细胞被抗原从Naive T细胞激活成癌细胞特异性T细胞之后,就可以去识别由各类抗原提呈细胞或者突变癌细胞所提呈的抗原多肽了。鉴于DC在癌细胞特异性T细胞初次激活中的核心功能,目前有一类癌症疫苗就是使用被抗原激活的DC回输患者作为治疗性疫苗,比如已经在美国上市的前列腺癌治疗性疫苗普罗文奇Provenge即是一款DC活细胞疫苗。但是DC细胞疫苗也面临着诸多问题,比如DC疫苗制备、储存、运输、使用过程中条件要求都很严格,DC疫苗作为活细胞疫苗易被各类微生物污染,而且活细胞疫苗一旦细胞发生基因污染或突变容易传导给机体内其他活细胞等等。除此之外,目前树突状细胞疫苗所负载的癌细胞抗原表位不够多样,也限制了树突状细胞疫苗的应用。因此,本发明旨在开发一种表面负载含有抗原提呈细胞细胞膜和与主要组织相容性复合物(MHC)分子结合后的抗原表位,且癌细胞抗原表位多样、既可以直接与T细胞结合激活Naive T细胞又可以通过被机体内的抗原提呈细胞吞噬间接激活Naive T细胞的、便于运输和储存的癌症疫苗。Cancer immunotherapy is one of the most important treatment methods for cancer, and cancer vaccine is one of the important methods of cancer immunotherapy. The usual mechanism of action of cancer vaccines is: the antigens loaded by cancer vaccines are phagocytosed and degraded by antigen-presenting cells, and the antigenic epitopes degraded into small peptides are combined with major histocompatibility complex (MHC) molecules and presented to the antigenic presentation cells. Present on the cell surface to activate cancer cell-specific T cells. The process of activating Naive T cells into cancer cell-specific T cells by cancer vaccines through antigen-presenting cells belongs to the initial activation of T cells, which requires dendritic cells (DC) to complete, and is completed in the lymph nodes. Once T cells are activated by antigens from Naive T cells to cancer cell-specific T cells, they can recognize antigenic polypeptides presented by various antigen-presenting cells or mutant cancer cells. In view of the core function of DCs in the primary activation of cancer cell-specific T cells, there is currently a type of cancer vaccine that uses antigen-activated DCs to infuse patients as therapeutic vaccines, such as Provenci, a prostate cancer therapeutic vaccine that has been marketed in the United States. Provenge is a DC live cell vaccine. However, DC cell vaccines also face many problems. For example, DC vaccines are subject to strict conditions during preparation, storage, transportation, and use. As live cell vaccines, DC vaccines are easily contaminated by various microorganisms, and once the cells of live cell vaccines are genetically contaminated Or the mutation is easily transmitted to other living cells in the body and so on. In addition, the antigenic epitopes of cancer cells loaded by current dendritic cell vaccines are not diverse enough, which also limits the application of dendritic cell vaccines. Therefore, the present invention aims to develop a surface-loaded antigenic epitope containing antigen-presenting cell membranes and binding to major histocompatibility complex (MHC) molecules, and the cancer cell antigenic epitopes are diverse, which can directly interact with T Cell binding activates Naive T cells, which in turn can indirectly activate Naive T cells by being phagocytosed by antigen-presenting cells in the body, which facilitates transportation and storage of cancer vaccines.
发明内容SUMMARY OF THE INVENTION
为解决上述技术问题,本发明提供了一种基于抗原提呈细胞膜组分的癌症疫苗。To solve the above technical problems, the present invention provides a cancer vaccine based on antigen-presenting cell membrane components.
本发明的第一个目的是提供一种基于抗原提呈细胞膜组分的癌症疫苗,该癌症疫苗包括以下其中之一:The first object of the present invention is to provide a cancer vaccine based on antigen-presenting cell membrane components, the cancer vaccine comprising one of the following:
(1)由激活的抗原提呈细胞细胞膜组分制备得到的纳米囊泡;(1) Nanovesicles prepared from activated antigen-presenting cell membrane components;
(2)内部和/或外部负载癌细胞全细胞组分,且表面负载激活的抗原提呈细胞细胞膜组分的第二递送粒子;(2) a second delivery particle that internally and/or externally loads whole cell components of cancer cells and loads activated antigen-presenting cell membrane components on its surface;
其中,in,
所述激活的抗原提呈细胞细胞膜组分由经预激活的抗原提呈细胞制备得到,所述预激活的抗原提呈细胞通过将抗原提呈细胞与负载肿瘤组织和/或癌细胞全细胞组分的第一递送粒子共孵育得到;The activated antigen-presenting cell membrane components are prepared from pre-activated antigen-presenting cells, which are prepared by combining antigen-presenting cells with tumor tissue and/or cancer cell whole cell groups. A fraction of the first delivery particles were co-incubated to obtain;
所述第一递送粒子和第二递送粒子(以下采用“递送粒子”表示第一递送粒子和/或第二递送粒子,第一递送粒子和第二递送粒子可同时经过或不经过以下处理过程,或其中之一经过以下处理过程)分别独立地为纳米粒子或微米粒子;所述抗原提呈细胞细胞膜组分为来自于细胞膜或细胞外囊泡膜的膜组分。The first delivery particle and the second delivery particle (hereinafter, "delivery particle" is used to refer to the first delivery particle and/or the second delivery particle, the first delivery particle and the second delivery particle may or may not be subjected to the following processes at the same time, Or one of them is nanoparticle or microparticle independently; the cell membrane component of antigen-presenting cell is the membrane component from cell membrane or extracellular vesicle membrane.
进一步地,癌细胞全细胞组分可为经癌细胞和/或肿瘤组织裂解得到的含有水溶性组分和非水溶性组分的全细胞组分,优选为肿瘤组织和/或癌细胞裂解后得到的全细胞组分所含有的全细胞抗原,该全细胞抗原包括经癌细胞和/或肿瘤组织裂解得到的水溶性抗原和非水溶性抗原,该非水溶性抗原经溶解剂溶解后负载于所述递送粒子上。Further, the whole cell fraction of cancer cells may be a whole cell fraction containing water-soluble components and water-insoluble components obtained by lysis of cancer cells and/or tumor tissue, preferably after lysis of tumor tissue and/or cancer cells. Whole cell antigens contained in the obtained whole cell component, the whole cell antigens include water-soluble antigens and water-insoluble antigens obtained by lysis of cancer cells and/or tumor tissues, and the water-insoluble antigens are dissolved in a lysing agent and then loaded on on the delivery particles.
进一步地,溶解剂选自尿素、盐酸胍、脱氧胆酸盐、十二烷基硫酸盐(如SDS)、甘油、蛋白质降解酶、白蛋白、卵磷脂、无机盐(0.1-2000mg/mL)、Triton、吐温、氨基酸、糖苷、胆碱中的至少一种。Further, the dissolving agent is selected from urea, guanidine hydrochloride, deoxycholate, dodecyl sulfate (such as SDS), glycerol, protein degrading enzymes, albumin, lecithin, inorganic salts (0.1-2000 mg/mL), At least one of Triton, Tween, amino acid, glycoside, choline.
进一步地,抗原提呈细胞可为树突状细胞、B细胞和/或巨噬细胞至少一种,优选地,可选择以上三种细胞中的两种,最优选为树突状细胞、B细胞和巨噬细胞的组合。Further, the antigen presenting cells can be at least one of dendritic cells, B cells and/or macrophages, preferably, two of the above three cells can be selected, most preferably dendritic cells, B cells and macrophages.
进一步地,所述递送粒子内部和/或外部还负载有免疫增强佐剂。Further, the inside and/or outside of the delivery particle is also loaded with an immune enhancing adjuvant.
进一步地,免疫增强佐剂包括但不限于微生物来源的免疫增强剂、人或动物免疫系统的产物、固有免疫激动剂、适应性免疫激动剂、化学合成药物、真菌多糖类、中药及其他类中的至少一类;免疫增强佐剂包括但不限于模式识别受体激动剂、卡介苗(BCG)、STING激动剂、卡介苗细胞壁骨架、卡介苗甲醇提取残余物、卡介苗胞壁酰二肽、草分枝杆菌、多抗甲素、矿物油、病毒样颗粒、免疫增强的再造流感病毒小体、霍乱肠毒素、皂苷及其衍生物、Resiquimod、胸腺素、新生牛肝活性肽、米喹莫特、多糖、姜黄素、STING激动剂、Toll样受体激动剂、免疫佐剂CpG、免疫佐剂poly(I:C)、免疫佐剂poly ICLC、短小棒状杆菌苗、溶血性链球菌制剂、辅酶Q10、左旋咪唑、聚胞苷酸、锰佐剂、铝佐剂、钙佐剂、各种细胞因子(如集落刺激因子)、白细胞介素、干扰素、聚肌苷酸、聚腺苷酸、明矾、磷酸铝、羊毛脂、角鲨烯、细胞因子、植物油、内毒素、脂质体佐剂、MF59、双链RNA、双链DNA、铝相关佐剂、CAF01、人参、黄芪的有效成分等。Further, immune-enhancing adjuvants include but are not limited to microbial-derived immune-enhancing agents, products of human or animal immune systems, innate immune agonists, adaptive immune agonists, chemically synthesized drugs, fungal polysaccharides, traditional Chinese medicines and others. At least one type of; immune-enhancing adjuvants include but are not limited to pattern recognition receptor agonists, Bacille Calmette-Guérin (BCG), STING agonists, BCG cell wall skeleton, BCG methanol extraction residue, BCG muramyl dipeptide, grass branch Bacillus, polyantibiotic A, mineral oil, virus-like particles, immune-enhancing reconstituted influenza virions, cholera enterotoxin, saponins and their derivatives, Resiquimod, thymosin, neonatal bovine liver active peptide, miquimod, polysaccharides , Curcumin, STING agonist, Toll-like receptor agonist, immune adjuvant CpG, immune adjuvant poly(I:C), immune adjuvant poly ICLC, Corynebacterium brevis vaccine, hemolytic streptococcus preparation, coenzyme Q10, Levamisole, polycytidylic acid, manganese adjuvant, aluminum adjuvant, calcium adjuvant, various cytokines (such as colony stimulating factor), interleukin, interferon, polyinosinic acid, polyadenylic acid, alum, Aluminum phosphate, lanolin, squalene, cytokines, vegetable oil, endotoxin, liposome adjuvant, MF59, double-stranded RNA, double-stranded DNA, aluminum-related adjuvant, CAF01, active ingredients of ginseng, astragalus, etc.
优选地,免疫增强佐剂为Toll样受体激动剂;更优选为两种及以上Toll样受体激动剂联用,保证纳米粒子或微米粒子被抗原提呈细胞吞噬后可以更好地激活癌细胞特异性T细胞。Preferably, the immune-enhancing adjuvant is a Toll-like receptor agonist; more preferably, two or more Toll-like receptor agonists are used in combination to ensure that the nanoparticles or microparticles can better activate cancer after being phagocytosed by antigen-presenting cells Cell-specific T cells.
进一步地,两种及以上Toll样受体激动剂联用为poly(I:C)/Poly(ICLC)与CpG-ODN(CpG寡脱氧核苷酸)联用。优选地,CpG-ODN为两种及以上的CpG-ODN。Further, the combination of two or more Toll-like receptor agonists is the combination of poly(I:C)/Poly(ICLC) and CpG-ODN (CpG oligodeoxynucleotide). Preferably, the CpG-ODNs are two or more CpG-ODNs.
进一步地,所述递送粒子内部和/或外部还负载有增加溶酶体逃逸的物质。Further, the inside and/or outside of the delivery particles are also loaded with substances that increase the escape of lysosomes.
进一步地,所述增加溶酶体逃逸的物质包括但不限于增加溶酶体内渗透压的载体材料、降低溶酶体膜稳定性的载体材料、具有质子海绵效应的物质。Further, the substances that increase lysosome escape include, but are not limited to, carrier materials that increase osmotic pressure in lysosomes, carrier materials that reduce lysosomal membrane stability, and substances that have proton sponge effect.
进一步地,增加溶酶体逃逸的物质包括但不限于氨基酸、聚氨基酸、有机高分子聚合物、核酸、多肽、脂类、糖类等。Further, substances that increase lysosome escape include but are not limited to amino acids, polyamino acids, organic polymers, nucleic acids, polypeptides, lipids, carbohydrates, and the like.
进一步地,所述递送粒子内部和/或外部还负载有主动靶向T细胞、抗原提呈细胞或NK细胞的靶头。靶头可为甘露糖、甘露聚糖、CD19抗体、CD3抗体、CD56抗体、CD20抗体、BCMA抗体、CD32抗体、CD11c抗体、CD103抗体、CD44抗体等。Further, the inside and/or outside of the delivery particle is also loaded with a target head that actively targets T cells, antigen-presenting cells or NK cells. The target head can be mannose, mannan, CD19 antibody, CD3 antibody, CD56 antibody, CD20 antibody, BCMA antibody, CD32 antibody, CD11c antibody, CD103 antibody, CD44 antibody and the like.
进一步地,上述基于抗原提呈细胞膜组分的癌症疫苗为冻干制剂,该冻干制剂由基于抗原提呈细胞膜组分的癌症疫苗经冷冻干燥制备得到,其中,冻干保护剂包括但不限于多羟基化合物、糖类和氨基酸。Further, the above-mentioned cancer vaccine based on the antigen-presenting cell membrane component is a freeze-dried preparation, and the freeze-dried preparation is prepared by freeze-drying the cancer vaccine based on the antigen-presenting cell membrane component, wherein the freeze-drying protective agent includes but is not limited to Polyols, carbohydrates and amino acids.
进一步地,多羟基化合物包括但不限于甘油、甘露醇、山梨醇、肌醇、硫醇、聚乙二醇等中的一种或多种。Further, polyhydroxy compounds include, but are not limited to, one or more of glycerol, mannitol, sorbitol, inositol, thiol, polyethylene glycol, and the like.
进一步地,糖类包括但不限于海藻糖、甘露糖、乳糖、蔗糖、葡聚糖、麦芽糖、菊糖、肝素等中的一种或多种。Further, carbohydrates include, but are not limited to, one or more of trehalose, mannose, lactose, sucrose, dextran, maltose, inulin, heparin, and the like.
进一步地,氨基酸包括但不限于精氨酸、组氨酸、酪氨酸、脯氨酸、色氨酸、甘氨酸等中的一种或多种。Further, amino acids include, but are not limited to, one or more of arginine, histidine, tyrosine, proline, tryptophan, glycine, and the like.
优选地,冻干保护剂为以下任意一种组合:Preferably, the lyoprotectant is any combination of the following:
按质量百分比计,By mass percentage,
(1)1~5%海藻糖、1~5%甘露醇和1~5%精氨酸,其余为水;(1) 1-5% trehalose, 1-5% mannitol and 1-5% arginine, and the rest is water;
(2)1~5%海藻糖、1~5%甘露醇和1~5%甘氨酸,其余为水;(2) 1-5% trehalose, 1-5% mannitol and 1-5% glycine, and the rest is water;
(3)1~5%海藻糖、1~5%甘露醇和1~5%赖氨酸,其余为水;(3) 1-5% trehalose, 1-5% mannitol and 1-5% lysine, and the rest is water;
(4)1~5%甘露醇、1~5%蔗糖和1~5%赖氨酸,其余为水;(4) 1-5% mannitol, 1-5% sucrose and 1-5% lysine, and the rest is water;
(5)1~5%海藻糖、1~5%甘露醇和1~5%明胶,其余为水;(5) 1-5% trehalose, 1-5% mannitol and 1-5% gelatin, and the rest is water;
(6)1~5%海藻糖、1~5%甘露醇和1~5%聚乙烯吡咯烷酮,其余为水;(6) 1-5% trehalose, 1-5% mannitol and 1-5% polyvinylpyrrolidone, and the rest is water;
(7)1~5%海藻糖、1~5%甘露醇和1~5%白蛋白,其余为水。(7) 1-5% trehalose, 1-5% mannitol and 1-5% albumin, and the rest is water.
进一步地,纳米粒子的粒径为1nm-1000nm;微米粒子的粒径为1μm-1000μm;纳米粒子或微米粒子表面为电中性,带负电或者带正电。Further, the particle size of the nanoparticle is 1 nm-1000 nm; the particle size of the microparticle is 1 μm-1000 μm; the surface of the nanoparticle or the microparticle is electrically neutral, negatively charged or positively charged.
进一步地,纳米粒子或微米粒子由有机合成高分子材料、天然高分子材料或无机材料制备得到,可以采用已有的制备方法制备得到,包括但不仅限于常见的溶剂挥发法、透析法、微流控法、挤出法、热熔法。Further, nanoparticles or micro-particles are prepared from organic synthetic polymer materials, natural polymer materials or inorganic materials, and can be prepared by existing preparation methods, including but not limited to common solvent evaporation methods, dialysis methods, microfluidics Control method, extrusion method, hot melt method.
进一步地,有机合成高分子材料包括PLGA、PLA、PGA、PEG、PCL、Poloxamer、PVA、PVP、PEI、PTMC、聚酸酐、PDON、PPDO、PMMA、聚氨基酸、合成多肽等;天然高分子材料包括卵磷脂、胆固醇、海藻酸盐、白蛋白、胶原蛋白、明胶、细胞膜成分、淀粉、糖类、多肽等;无机材料包括三氧化二铁、四氧化三铁、碳酸盐、磷酸盐等。Further, organic synthetic polymer materials include PLGA, PLA, PGA, PEG, PCL, Poloxamer, PVA, PVP, PEI, PTMC, polyanhydrides, PDON, PPDO, PMMA, polyamino acids, synthetic polypeptides, etc.; natural polymer materials include Lecithin, cholesterol, alginate, albumin, collagen, gelatin, cell membrane components, starch, carbohydrates, polypeptides, etc.; inorganic materials include ferric oxide, ferric tetroxide, carbonate, phosphate, etc.
进一步地,水溶性抗原或非水溶性抗原负载于纳米粒子或微米粒子表面的方式包括吸附、共价连接、电荷相互作用、疏水相互作用、一步或多步的固化、矿化和包裹中的至少一种。Further, the manner in which water-soluble antigens or water-insoluble antigens are loaded on the surface of nanoparticles or microparticles includes at least one of adsorption, covalent attachment, charge interaction, hydrophobic interaction, one-step or multi-step solidification, mineralization, and encapsulation. A sort of.
进一步地,纳米粒子或微米粒子在制备过程中可以不做修饰处理,也可以采用适当的修饰技术以提高纳米粒子或微米粒子的抗原负载量。修饰技术包括但不限于生物矿化(如硅化、钙化、镁化)、凝胶化、交联、化学修饰、添加带电物质等。Further, the nanoparticles or microparticles may not be modified during the preparation process, and appropriate modification techniques may also be used to increase the antigen loading of the nanoparticles or microparticles. Modification techniques include, but are not limited to, biomineralization (eg, silicidation, calcification, magnesia), gelation, cross-linking, chemical modification, addition of charged species, and the like.
进一步地,抗原被负载于纳米粒子或微米粒子内部的形式为任何可以将其负载于纳米粒子或微米粒子内部的方式,如包载。Further, the form in which the antigen is loaded inside the nanoparticle or the microparticle can be any manner in which the antigen can be loaded inside the nanoparticle or the microparticle, such as encapsulation.
进一步地,抗原被负载于纳米粒子或微米粒子表面的方式包括但不限于吸附、共价连接、电荷相互作用(如添加带正电的物质、添加带负电的物质)、疏水相互作用、一步或多步的固化、矿化、包裹等。Further, the manner in which the antigen is loaded on the surface of nanoparticles or microparticles includes, but is not limited to, adsorption, covalent attachment, charge interaction (such as adding a positively charged substance, adding a negatively charged substance), hydrophobic interaction, one-step or Multi-step curing, mineralization, wrapping, etc.
进一步地,负载于纳米粒子或微米粒子表面的水溶性抗原和/或非水溶性抗原负载后为一层或多层,表面负载多层水溶性抗原和/或非水溶性抗原时,层与层之间为修饰物。Further, the water-soluble antigens and/or water-insoluble antigens loaded on the surface of nanoparticles or microparticles are one or more layers, and when the surface is loaded with multi-layers of water-soluble antigens and/or water-insoluble antigens, the layers are In between are modifiers.
本发明的第二个目的是提供上述基于抗原提呈细胞膜组分的癌症疫苗的制备方法,包括以下步骤:The second object of the present invention is to provide the above-mentioned preparation method of the cancer vaccine based on the antigen-presenting cell membrane component, comprising the following steps:
S1、将抗原提呈细胞与负载癌细胞全细胞组分的第一递送粒子共孵育以激活抗原提呈细胞;S1, co-incubating the antigen-presenting cells with the first delivery particles loaded with the whole cell component of the cancer cells to activate the antigen-presenting cells;
S2、将S1中激活的抗原提呈细胞的细胞膜组分制备成纳米囊泡,得到所述基于抗原提呈细胞膜组分的癌症疫苗;S2, preparing the cell membrane components of the antigen-presenting cells activated in S1 into nanovesicles to obtain the cancer vaccine based on the antigen-presenting cell membrane components;
或获取S1中激活的抗原提呈细胞的细胞膜组分,将所述细胞膜组分和/或纳米囊泡与负载癌细胞全细胞组分的第二递送粒子共作用,得到所述基于抗原提呈细胞膜组分的癌症疫苗。Or obtaining the cell membrane fraction of the antigen-presenting cells activated in S1, and co-acting the cell membrane fraction and/or nanovesicles with the second delivery particle loaded with the cancer cell whole cell fraction to obtain the antigen-presenting-based Cancer vaccines with cell membrane components.
进一步地,获取S1中激活的抗原提呈细胞的细胞膜组分的方式可为机械破坏、膜过滤、梯度离心或化学处理;共作用的方式为共孵育、共挤出、超声、搅拌、均质化或匀浆。Further, the method of obtaining the cell membrane components of the activated antigen-presenting cells in S1 can be mechanical destruction, membrane filtration, gradient centrifugation or chemical treatment; the method of co-action is co-incubation, co-extrusion, ultrasound, stirring, homogenization. slurried or homogenized.
进一步地,梯度离心为离心速度依次增加的梯度离心;膜过滤时所使用的滤膜的孔径依次由大到小;机械破坏的方式可为超声、均质化、匀浆、高速搅拌、高压破坏、高剪切力破坏、溶胀;化学处理的方式可为将细胞置于含有化学物质的低渗透压的PBS、葡萄糖溶液或盐溶液、皱缩等。其中,超声为低功率超声(低于500W)。Further, gradient centrifugation is gradient centrifugation in which the centrifugal speed increases in turn; the pore size of the filter membrane used in membrane filtration is sequentially from large to small; the way of mechanical destruction can be ultrasonic, homogenization, homogenization, high-speed stirring, high-pressure destruction. , high shear damage, swelling; chemical treatment methods can be placed in PBS, glucose solution or saline solution containing chemical substances with low osmotic pressure, shrinkage, etc. Among them, the ultrasound is low-power ultrasound (below 500W).
进一步地,共孵育至少4小时,使抗原能够递送到抗原提呈细胞内,且可被抗原提呈细胞处理和提呈到抗原提呈细胞表面。Further, co-incubation for at least 4 hours enables the antigen to be delivered into the antigen-presenting cells, processed by the antigen-presenting cells and presented to the surface of the antigen-presenting cells.
进一步地,共孵育的过程中孵育液中含有细胞因子和/或抗体。Further, the incubation solution contains cytokines and/or antibodies during the co-incubation.
进一步地,细胞因子选自白介素、肿瘤坏死因子、干扰素、生长因子、集落刺激因子中的至少一种。Further, the cytokine is selected from at least one of interleukin, tumor necrosis factor, interferon, growth factor and colony stimulating factor.
进一步地,细胞因子包括但不限于白介素2(IL-2)、白介素7(IL-7)、白介素14(IL-14)、白介素4(IL-4)、白介素15(IL-15)、白介素21(IL-21)、白介素17(IL-17)、白介素12(IL-12)、白介素6(IL-6)、白介素33(IL-33)、γ干扰素(IFN-γ)、TNF-α、粒细胞-巨噬细胞集落刺激因子(GM-CSF)、巨噬细胞集落刺激因子(M-CSF)等。Further, cytokines include but are not limited to interleukin 2 (IL-2), interleukin 7 (IL-7), interleukin 14 (IL-14), interleukin 4 (IL-4), interleukin 15 (IL-15), interleukin 21 (IL-21), Interleukin 17 (IL-17), Interleukin 12 (IL-12), Interleukin 6 (IL-6), Interleukin 33 (IL-33), Interferon gamma (IFN-γ), TNF- α, granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), etc.
进一步地,抗体包括但不限于αCD-3抗体、αCD-4抗体、αCD-8抗体、αCD-28抗体、CD-40抗体、CD-80抗体、αOX-40抗体、αOX-40L抗体、CD86抗体等。Further, antibodies include but are not limited to αCD-3 antibody, αCD-4 antibody, αCD-8 antibody, αCD-28 antibody, CD-40 antibody, CD-80 antibody, αOX-40 antibody, αOX-40L antibody, CD86 antibody Wait.
优选地,孵育过程中加入以下任意一种组合:Preferably, any one of the following combinations is added during the incubation:
(1)粒细胞-巨噬细胞集落刺激因子、白介素2、白介素7和白介素12;(1) granulocyte-macrophage colony-stimulating factor, interleukin-2, interleukin-7 and interleukin-12;
(2)粒细胞-巨噬细胞集落刺激因子、白介素4和肿瘤坏死因子α;(2) granulocyte-macrophage colony-stimulating factor, interleukin-4 and tumor necrosis factor alpha;
(3)粒细胞-巨噬细胞集落刺激因子、白介素2、白介素7、白介素12、CD80抗体和CD40抗体;(3) granulocyte-macrophage colony-stimulating factor, interleukin-2, interleukin-7, interleukin-12, CD80 antibody and CD40 antibody;
(4)粒细胞-巨噬细胞集落刺激因子、白介素12和CD80抗体;(4) granulocyte-macrophage colony-stimulating factor, interleukin-12 and CD80 antibodies;
(5)白介素2、白介素7和白介素15;(5)
(6)白介素4、白介素6和白介素10;(6)
(7)粒细胞-巨噬细胞集落刺激因子、白介素2、白介素7、白介素12和CD86抗体;(7) granulocyte-macrophage colony-stimulating factor, interleukin-2, interleukin-7, interleukin-12 and CD86 antibodies;
(8)粒细胞-巨噬细胞集落刺激因子、白介素2、白介素7、白介素12、白蛋白和CD80抗体;(8) granulocyte-macrophage colony-stimulating factor, interleukin-2, interleukin-7, interleukin-12, albumin and CD80 antibodies;
(9)粒细胞-巨噬细胞集落刺激因子、白介素2、白介素7、白介素12和CD40抗体;(9) granulocyte-macrophage colony-stimulating factor, interleukin-2, interleukin-7, interleukin-12 and CD40 antibodies;
(10)粒细胞-巨噬细胞集落刺激因子、巨噬细胞集落刺激因子、白介素2、白介素7、白介素12、γ干扰素和CD80抗体;(10) granulocyte-macrophage colony-stimulating factor, macrophage colony-stimulating factor,
(11)粒细胞-巨噬细胞集落刺激因子、白介素2、白介素7、白介素12、γ干扰素、CD80抗体和CD40抗体。(11) granulocyte-macrophage colony-stimulating factor, interleukin-2, interleukin-7, interleukin-12, gamma interferon, CD80 antibody and CD40 antibody.
进一步地,在步骤S1中,还包括将激活的抗原提呈细胞进行洗涤的过程,洗涤过称中使用的洗涤液中可含有蛋白酶抑制剂和/或磷酸酶抑制剂。Further, in step S1, a process of washing the activated antigen-presenting cells is also included, and the washing solution used in the washing process may contain protease inhibitors and/or phosphatase inhibitors.
树突状细胞癌症疫苗是癌症疫苗的一种,现有技术中,由于激活方式的限制,树突状细胞(DC)所能负载的抗原相对有限,因而注入人体后T细胞被激活的种类和克隆数很少,而且,由于DC属于活细胞制品,作为疫苗使用的被激活的DC在储存、运输和给药过程都存在诸多缺陷。本发明中,先使用负载癌细胞抗原的纳米粒子和/或微米粒子激活抗原提呈细胞,再将抗原提呈细胞经过一定方法处理制备成了纳米疫苗或微米疫苗,所得疫苗负载有癌细胞全细胞组分抗原,因而注射进入人体后可以激活广谱多样的癌细胞特异性T细胞,从而可以更多和更好的识别和杀灭癌细胞。Dendritic cell cancer vaccine is a kind of cancer vaccine. In the prior art, due to the limitation of activation mode, dendritic cells (DC) can carry relatively limited antigens, so the types of T cells activated after injection into the human body and The number of clones is small, and since DCs are live cell preparations, activated DCs used as vaccines have many defects in storage, transportation and administration. In the present invention, the antigen-presenting cells are first activated by using nano-particles and/or micro-particles loaded with cancer cell antigens, and then the antigen-presenting cells are processed by a certain method to prepare a nano-vaccine or a micro-vaccine, and the obtained vaccine is loaded with cancer cell whole Therefore, after injection into the human body, a broad spectrum of cancer cell-specific T cells can be activated, so that more and better cancer cells can be identified and killed.
而且,由于本发明所述纳米疫苗或微米疫苗表面负载细胞膜组分且细胞膜上含有与MHC结合后的广谱多肽抗原,所以该纳米疫苗或微米疫苗可以作为人工抗原提呈细胞直接与T细胞作用将其激活为癌细胞特异性T细胞;此外纳米疫苗或微米疫苗还可以被抗原提呈细胞吞噬,进而通过抗原提呈细胞再间接激活癌细胞特异性T细胞。现有的疫苗只能通过抗原提呈细胞间接激活T细胞,而本发明所述纳米疫苗或微米疫苗可以通过直接激活T细胞和通过抗原提呈细胞间接激活T细胞两种方式激活T细胞。因此本发明基于上述癌症疫苗,提供了一种使用抗原提呈细胞来源的癌症疫苗直接激活T细胞的方法,将癌症疫苗与T细胞在体外共孵育以诱导激活得到癌细胞特异性T细胞。Moreover, since the surface of the nano-vaccine or micro-vaccine of the present invention is loaded with cell membrane components and the cell membrane contains broad-spectrum polypeptide antigens combined with MHC, the nano-vaccine or micro-vaccine can be used as artificial antigen-presenting cells to directly interact with T cells activate them into cancer cell-specific T cells; in addition, nano-vaccine or micro-vaccine can also be phagocytosed by antigen-presenting cells, and then indirectly activate cancer-cell-specific T cells through antigen-presenting cells. Existing vaccines can only activate T cells indirectly through antigen-presenting cells, while the nano-vaccine or micro-vaccine of the present invention can activate T cells in two ways: direct activation of T cells and indirect activation of T cells through antigen-presenting cells. Therefore, based on the above-mentioned cancer vaccine, the present invention provides a method for directly activating T cells using antigen-presenting cell-derived cancer vaccine, and co-incubating the cancer vaccine and T cells in vitro to induce activation to obtain cancer cell-specific T cells.
本发明的第三个目的是提供上述基于抗原提呈细胞膜组分的癌症疫苗在制备治疗或预防癌症药物中的应用。The third object of the present invention is to provide the application of the above-mentioned antigen-presenting cell membrane component-based cancer vaccine in the preparation of a drug for treating or preventing cancer.
借由上述方案,本发明至少具有以下优点:By means of the above scheme, the present invention has at least the following advantages:
(1)本发明可以将多种抗原提呈细胞的膜组分、负载的MHC分子、抗原整合到一个纳米疫苗或微米疫苗内,因此,一个疫苗内可以含有包括DC细胞、B细胞或巨噬细胞等多种抗原提呈细胞的膜组分,因而一个纳米疫苗粒子同时可以具有多种抗原提呈细胞的功能和优势。(1) The present invention can integrate membrane components, loaded MHC molecules, and antigens of various antigen-presenting cells into a nano-vaccine or micro-vaccine. Therefore, a vaccine can contain DC cells, B cells or macrophages. The membrane components of various antigen-presenting cells such as cells, so a nano-vaccine particle can simultaneously have the functions and advantages of multiple antigen-presenting cells.
(2)本发明的疫苗中可以含有包括DC细胞在内多种抗原提呈细胞的膜组分,抗原提呈细胞细胞膜上的蛋白质对于淋巴结以及淋巴结内的免疫细胞会具有特殊的亲和力,因而注射进入体内后具有归巢淋巴结的特性,具有天然靶向淋巴结的功效能更好的激活癌细胞特异性免疫反应。(2) The vaccine of the present invention can contain membrane components of various antigen-presenting cells including DC cells. The proteins on the cell membrane of antigen-presenting cells have special affinity for lymph nodes and immune cells in the lymph nodes. After entering the body, it has the characteristics of homing to the lymph nodes, and the effect of naturally targeting the lymph nodes can better activate the specific immune response of cancer cells.
(3)本发明使用负载癌细胞抗原的纳米粒子和/或微米粒子激活抗原提呈细胞,再使用抗原提呈细胞制备成疫苗,因而所制备的纳米疫苗可以负载所制备纳米粒子和/或微米粒子所负载的全部抗原组分,纳米疫苗可以负载广谱多样的癌细胞特异性抗原。(3) The present invention uses nanoparticles and/or microparticles loaded with cancer cell antigens to activate antigen-presenting cells, and then uses antigen-presenting cells to prepare a vaccine, so the prepared nanovaccine can be loaded with the prepared nanoparticles and/or microparticles With all the antigen components carried by the particles, the nanovaccine can carry a broad spectrum of cancer cell-specific antigens.
(4)本发明的疫苗来源于微米大小的抗原提呈细胞,所有组分均是生物相容性和可降解的,安全性好,同时解决了活细胞疫苗在储存、运输和注射时条件要求苛刻的问题,而且疗效比活细胞疫苗更好。(4) The vaccine of the present invention is derived from micron-sized antigen-presenting cells, and all components are biocompatible and degradable, with good safety, and at the same time, the requirements for storage, transportation and injection of live cell vaccines are solved. Tough question and better efficacy than live cell vaccines.
上述说明仅是本发明技术方案的概述,为了能够更清楚了解本发明的技术手段,并可依照说明书的内容予以实施,以下以本发明的较佳实施例并配合详细附图说明如后。The above description is only an overview of the technical solution of the present invention. In order to understand the technical means of the present invention more clearly and implement it according to the content of the description, the following description is given with the preferred embodiments of the present invention and the detailed drawings.
附图说明Description of drawings
为了使本发明的内容更容易被清楚的理解,下面根据本发明的具体实施例并结合附图,对本发明作进一步详细的说明。In order to make the content of the present invention easier to understand clearly, the present invention will be described in further detail below according to specific embodiments of the present invention and in conjunction with the accompanying drawings.
图1为本发明纳米疫苗或微米疫苗的制备过程及应用示意图;其中,a为水溶性抗原和非水溶性抗原分别收集和制备纳米粒子或微米粒子的示意图;b为采用含有溶解剂的溶解液溶解癌细胞全细胞抗原和制备纳米粒子或微米粒子的示意图;c为使用a或b中制备的上述粒子激活抗原提呈细胞后,将抗原提呈细胞制备成纳米疫苗或微米疫苗,并用该类纳米疫苗或微米疫苗预防或治疗癌症的示意图;Fig. 1 is the preparation process and application schematic diagram of nano-vaccine or micro-vaccine of the present invention; Wherein, a is the schematic diagram of the collection and preparation of nano-particles or micro-particles for water-soluble antigens and water-insoluble antigens respectively; b is the use of a lysing solution containing a dissolving agent Schematic diagram of dissolving whole cell antigens of cancer cells and preparing nanoparticles or microparticles; c is the use of the above-mentioned particles prepared in a or b to activate antigen-presenting cells, and then prepare the antigen-presenting cells into nano-vaccine or micro-vaccine, and use this type of vaccine Schematic diagram of nano-vaccine or micro-vaccine to prevent or treat cancer;
图2-20分别为实施例1-19中用纳米疫苗或微米疫苗预防或治疗癌症时小鼠肿瘤生长速度和生存期实验结果;a,预防或治疗癌症时的肿瘤生长速度实验结果(n≥8);b,预防或治疗癌症时的小鼠生存期实验结果(n≥8),每个数据点为平均值±标准误差(mean±SEM);c和d为使用流式细胞术分析纳米疫苗激活的CD8+和CD4+的癌细胞特异性T细胞占总的CD8+和CD4+T细胞的比例;e为纳米疫苗透射电镜(TEM)扫描图像;a图中肿瘤生长抑制实验的显著性差异采用ANOVA法分析,b图中显著性差异采用Kaplan-Meier和log-rank test分析;***表示与PBS空白对照组相比p<0.005,有显著性差异;**表示与PBS空白对照组相比p<0.01,有显著性差异;代表与纳米粒/微米粒激活抗原提呈细胞过程中加入特定细胞因子共孵育后所制备的抗原提呈细胞的纳米疫苗对照组相比p<0.05,有显著性差异;&&&表示与空白纳米粒/微米粒+游离裂解液激活的抗原提呈细胞制备的纳米疫苗对照组相比p<0.005,有显著性差异;&&表示与空白纳米粒/微米粒+游离裂解激活的抗原提呈细胞制备的纳米疫苗对照组相比p<0.01,有显著性差异;δδδ代表与多肽纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗相比p<0.005,有显著性差异;δδ代表与多肽纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗组相比p<0.01,有显著性差异;ΔΔ代表与使用被负载全细胞组分的纳米粒子激活的混合抗原提呈细胞活细胞疫苗组相比p<0.01,有显著性差异;★代表与使用1%海藻糖+2%甘露醇+2%PVP水溶液作为冻干保护剂冷冻干燥制备的纳米疫苗或微米疫苗相比p<0.05,有显著性差异;Ψ代表与纳米粒/微米粒激活的DC+B细胞所制备的纳米疫苗组相比p<0.05,有显著性差异;Ω代表与纳米粒/微米粒激活的B细胞所制备的纳米疫苗组相比p<0.05,有显著性差异;χχ代表与使用特定细胞因子组合辅助微米粒子激活的抗原提呈细胞制备的纳米疫苗或微米疫苗相比p<0.01,有显著性差异;ω代表与只是内部负载癌细胞全细胞组分但是表面不负载被激活的抗原提呈细胞细胞膜组分的纳米疫苗或微米疫苗相比p<0.05,有显著性差异;Μ代表与纳米粒/微米粒激活的巨噬细胞所制备的纳米疫苗组相比p<0.05,有显著性差异;θθθ代表与不负载佐剂的纳米粒/微米粒辅助分离的T细胞组相比p<0.005,有显著性差异;θθ代表与不负载佐剂的纳米粒/微米粒辅助分离的T细胞组相比p<0.01,有显著性差异;ππ代表与不负载溶酶体逃逸物质的纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗组相比p<0.01,有显著性差异;π代表与不负载溶酶体逃逸物质的纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗组相比p<0.05,有显著性差异;ξξ代表与只负载一种CpG+Poly(I:C)混合佐剂的纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗相比p<0.01,有显著性差异;ΣΣ代表与只使用抗原提呈细胞细胞膜组分制备的纳米疫苗或微米疫苗相比p<0.01,有显著性差异;β代表与使用2%蔗糖+2%甘露醇+1%赖氨酸水溶液作为冻干保护剂冷冻干燥制备的纳米疫苗或微米疫苗相比p<0.05,有显著性差异;ξ代表与只负载一种CpG+Poly(I:C)混合佐剂的纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗相比p<0.05,有显著性差异;μ代表与使用两种A类CpG与toll样受体3混合佐剂的纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗组相比p<0.05,有显著性差异;ρ代表与只负载一种佐剂(两类CpG)的纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗组相比p<0.05,有显著性差异;τ代表与纳米粒/微米粒激活抗原提呈细胞过程中不加入IL-2和IL-7共孵育所制备纳米疫苗相比p<0.01,有显著性差异;###表示与未被任何纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗对照组相比p<0.005,有显著性差异;##表示与未被任何纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗对照组相比p<0.01,有显著性差异;#表示与未被任何纳米粒/微米粒激活的抗原提呈细胞制备的纳米疫苗对照组相比p<0.05,有显著性差异。Figures 2-20 are respectively the experimental results of tumor growth rate and survival period of mice when using nano-vaccine or micro-vaccine to prevent or treat cancer in Example 1-19; a, experimental results of tumor growth rate when preventing or treating cancer (n≥ 8); b, the experimental results of mouse survival when preventing or treating cancer (n ≥ 8), each data point is the mean ± standard error (mean ± SEM); c and d are the analysis of nanometers using flow cytometry The proportion of vaccine-activated CD8 + and CD4 + cancer cell-specific T cells to total CD8 + and CD4 + T cells; e is the transmission electron microscope (TEM) scan image of the nanovaccine; the significance of the tumor growth inhibition experiment in a The difference was analyzed by ANOVA method, and the significant difference in figure b was analyzed by Kaplan-Meier and log-rank test; *** means p<0.005 compared with PBS blank control group, there is a significant difference; ** means compared with PBS blank control group Compared with the group, p<0.01, there is a significant difference; Represents the nanovaccine control group prepared by adding specific cytokines in the process of activating antigen-presenting cells by nanoparticles/microparticles compared with the nano-vaccine control group, p<0.05, there is a significant difference; &&& means that with blank nanoparticles Compared with the control group, the nano-vaccine prepared by /microparticles + free lysate activated antigen-presenting cells has a significant difference, p<0.005; Compared with the nanovaccine control group, p<0.01, there is a significant difference; δδδ represents p<0.005 compared with the nanovaccine prepared by the antigen-presenting cells activated by polypeptide nanoparticles/microparticles; Compared with the nanovaccine group prepared by the APCs activated by particles/microparticles, p<0.01, there is a significant difference; Compared with p < 0.01, there is a significant difference; ★ means p < 0.05 compared with the nano-vaccine or micro-vaccine prepared by freeze-drying using 1% trehalose + 2% mannitol + 2% PVP aqueous solution as freeze-drying protective agent. Significant difference; Ψ represents p<0.05 compared with the nanovaccine group prepared by nanoparticle/microparticle activated DC+B cells; Ω represents the nanovaccine group prepared with nanoparticle/microparticle activated B cells Compared with the nanovaccine group, p<0.05, there is a significant difference; χχ represents a significant difference compared with p<0.01 compared with the nanovaccine or microvaccine prepared from antigen-presenting cells activated by microparticles using a specific combination of cytokines; ω Represents a significant difference compared with the nano-vaccine or micro-vaccine that only loads the whole cell components of cancer cells internally but does not load the activated antigen-presenting cell membrane components on the surface; M represents a significant difference with nanoparticles/microparticles Compared with the nanovaccine group prepared by activated macrophages, p<0.05, there is a significant difference; θθθ represents p<0.005 compared with the T cell group assisted by the separation of nanoparticles/microparticles without adjuvant, and there is a significant difference Difference; θθ represents p<0.01, significant difference compared with unadjuvanted nano/microparticle-assisted isolated T cell group; Compared with the nanovaccine group prepared by antigen-presenting cells, p<0.01, there is a significant difference; <0.05, there is a significant difference; ξξ represents that compared with the nanovaccine prepared by nanoparticle/microparticle activated antigen-presenting cells loaded with only one CpG+Poly(I:C) mixed adjuvant, p<0.01, there is a significant difference Sexual differences; ΣΣ represents p<0.01 compared with nano-vaccine or micro-vaccine prepared by using only antigen-presenting cell membrane fractions; Nanoparticles prepared by freeze-drying of aqueous acid solution as lyoprotectant Compared with vaccine or microvaccine, p<0.05, there is a significant difference; ξ represents the phase of nanovaccine prepared with nanoparticle/microparticle activated antigen-presenting cells loaded with only one CpG+Poly(I:C) mixed adjuvant Ratio p<0.05, there is a significant difference; μ represents p<0.05 compared with the nanovaccine group prepared by nanoparticles/microparticle-activated antigen-presenting cells using two kinds of A-class CpG and toll-like receptor 3 mixed adjuvants , there is a significant difference; ρ represents a significant difference compared with the nanovaccine group prepared by nanoparticle/microparticle activated antigen-presenting cells loaded with only one adjuvant (two types of CpG); p<0.05, a significant difference; τ represents Compared with the nanovaccine prepared by co-incubating with nanoparticles/microparticles without adding IL-2 and IL-7 in the process of activating antigen-presenting cells, there is a significant difference p<0.01; Compared with the nanovaccine control group prepared by the antigen-presenting cells activated by microparticles, there is a significant difference, p<0.005; Ratio p<0.01, there is a significant difference; # means p<0.05, a significant difference compared with the nanovaccine control group prepared from antigen-presenting cells not activated by any nanoparticles/microparticles.
具体实施方式Detailed ways
下面结合附图和具体实施例对本发明作进一步说明,以使本领域的技术人员可以更好地理解本发明并能予以实施,但所举实施例不作为对本发明的限定。The present invention will be further described below with reference to the accompanying drawings and specific embodiments, so that those skilled in the art can better understand the present invention and implement it, but the embodiments are not intended to limit the present invention.
本发明所述的用于预防或治疗癌症的基于抗原提呈细胞膜组分的癌症疫苗,其包含由被负载癌细胞和/或肿瘤组织全细胞组分的纳米粒子和/或微米粒子激活的抗原提呈细胞制备所得的纳米疫苗或微米疫苗。纳米粒子和/或微米粒子负载癌细胞全细胞组分或其混合物。全细胞组分中含有全细胞抗原。制备预防或治疗癌症的纳米疫苗,其制备过程及应用领域如图1所示。The cancer vaccine based on antigen-presenting cell membrane components for preventing or treating cancer according to the present invention comprises an antigen activated by nanoparticles and/or microparticles loaded with cancer cells and/or tumor tissue whole cell components Present the nanovaccine or microvaccine prepared by the cells. Nanoparticles and/or microparticles are loaded with cancer cell whole cell components or mixtures thereof. Whole cell antigens are contained in the whole cell fraction. The preparation process and application fields of nano-vaccine for preventing or treating cancer are shown in Fig. 1 .
在制备用于激活抗原提呈细胞的负载癌细胞和/或肿瘤组织全细胞组分的纳米粒子或微米粒子时,可裂解细胞或组织后先分别收集水溶性抗原和水不溶性抗原并分别制备纳米或微米粒子系统;或者也可以直接采用含有溶解剂的溶解液直接裂解细胞或组织并溶解癌细胞全细胞抗原并制备纳米或微米粒子系统。本发明所述癌细胞全细胞抗原在裂解前或(和)裂解后既可经过包括但不限于灭活或(和)变性、固化、生物矿化、离子化、化学修饰、核酸酶处理等处理后再制备纳米粒子或微米粒子;也可细胞裂解前或(和)裂解后不经过任何灭活或(和)变性、固化、生物矿化、离子化、化学修饰、核酸酶处理直接制备纳米粒子或微米粒子。本发明部分实施例中,肿瘤组织细胞在裂解前经过了灭活或(和)变性处理,在实际使用过程中也可以在细胞裂解后做灭活或(和)变性处理,或者也可以细胞裂解前和裂解后均做灭活或(和)变性处理;本发明部分实施例中细胞裂解前或(和)裂解后的灭活或(和)变性处理方法为紫外照射和高温加热,在实际使用过程中也可以采用包括但不限于放射线辐照、高压、固化、生物矿化、离子化、化学修饰、核酸酶处理、胶原酶处理、冷冻干燥等处理方法。本领域技术人员可以理解,在实际应用过程中技术人员可根据具体情况进行适当调整。When preparing nanoparticles or microparticles loaded with cancer cells and/or tumor tissue whole cell components for activating antigen-presenting cells, the cells or tissues can be lysed and then water-soluble antigens and water-insoluble antigens can be collected and prepared separately. Or a microparticle system; or a lysing solution containing a lysing agent can be used to directly lyse cells or tissues and lyse whole cell antigens of cancer cells to prepare a nanoparticle or microparticle system. The cancer cell whole cell antigen of the present invention can be processed before or (and) after lysis, including but not limited to inactivation or (and) denaturation, solidification, biomineralization, ionization, chemical modification, nuclease treatment, etc. Nanoparticles or microparticles are then prepared; nanoparticles can also be directly prepared before or (and) after cell lysis without any inactivation or (and) denaturation, solidification, biomineralization, ionization, chemical modification, and nuclease treatment. or micron particles. In some embodiments of the present invention, tumor tissue cells are inactivated or (and) denatured before lysis, and in actual use, inactivation or (and) denaturation can also be performed after cell lysis, or cell lysis can also be performed. Before and after lysis, inactivation or (and) denaturation treatment is performed; in some embodiments of the present invention, the inactivation or (and) denaturation treatment methods before or (and) after lysis of cells are ultraviolet irradiation and high temperature heating. In the process, treatment methods including but not limited to radiation irradiation, high pressure, solidification, biomineralization, ionization, chemical modification, nuclease treatment, collagenase treatment, freeze-drying and the like can also be used. Those skilled in the art can understand that in the actual application process, the skilled person can make appropriate adjustments according to specific conditions.
在使用纳米粒子或微米粒子体外激活抗原提呈细胞时,可以使用细胞因子和/或抗体辅助提高激活效率,抗原提呈细胞可以来源于自体或者同种异体,也可以来自于细胞系或者干细胞。抗原提呈细胞可以是DC细胞、B细胞、巨噬细胞或者上述三者的任意混合物,也可以是其他具有抗原提呈功能的细胞。When using nanoparticles or microparticles to activate antigen-presenting cells in vitro, cytokines and/or antibodies can be used to help improve the activation efficiency. Antigen-presenting cells can be derived from autologous or allogeneic cells, or from cell lines or stem cells. The antigen presenting cells can be DC cells, B cells, macrophages or any mixture of the above three, and can also be other cells with antigen presenting function.
在抗原提呈细胞被激活后,使用低频率超声和梯度离心等方法将抗原提呈细胞制备成纳米大小的纳米疫苗或微米疫苗。在实际制备是也可以采用其他可以将微米大小的活的抗原提呈细胞制备成纳米大小或微米大小的不具有细胞活性的纳米疫苗或微米疫苗的方法。After the antigen-presenting cells are activated, low-frequency ultrasound and gradient centrifugation are used to prepare the antigen-presenting cells into nano-sized nano-vaccine or micro-vaccine. In actual preparation, other methods for preparing micron-sized live antigen-presenting cells into nano-sized or micron-sized nano-vaccine or micro-vaccine without cellular activity can also be used.
在一些实施方案中,采用负载癌细胞全细胞抗原的纳米粒子或微米粒子激活的抗原提呈细胞制备纳米疫苗或微米疫苗的具体制备方法如下:In some embodiments, the specific preparation method for preparing nano-vaccine or micro-vaccine using antigen-presenting cells activated by nanoparticles or micro-particles loaded with cancer cell whole cell antigens is as follows:
步骤1,将第一预定体积的含有第一预定浓度的水相溶液加入第二预定体积的含有第二预定浓度制备粒子原材料的有机相中。
在一些实施例中,水相溶液可含有癌细胞裂解物中的各组分以及免疫增强佐剂;癌细胞裂解物中的各组分在制备时分别为水溶性抗原或是溶于尿素或盐酸胍等溶解剂中的原非水溶性抗原。水相溶液所含有的水溶性抗原的浓度或原非水溶性抗原的浓度,也即第一预定浓度要求蛋白质多肽浓度含量大于1ng/mL,能负载足够癌细胞全细胞抗原以激活相关细胞。免疫增强佐剂在初始水相中的浓度为大于0.01ng/mL。In some embodiments, the aqueous phase solution may contain each component in the cancer cell lysate and an immune-enhancing adjuvant; each component in the cancer cell lysate is prepared as a water-soluble antigen or soluble in urea or hydrochloric acid, respectively. Originally water-insoluble antigens in solubilizing agents such as guanidine. The concentration of the water-soluble antigen contained in the aqueous solution or the concentration of the original water-insoluble antigen, that is, the first predetermined concentration requires that the protein polypeptide concentration is greater than 1 ng/mL, which can load enough cancer cell whole cell antigens to activate relevant cells. The concentration of the immunopotentiating adjuvant in the initial aqueous phase was greater than 0.01 ng/mL.
在一些实施例中,水相溶液含有肿瘤组织裂解物中的各组分以及免疫增强佐剂;肿瘤组织裂解物中的各组分在制备时分别为水溶性抗原或者是溶于尿素或盐酸胍等溶解剂中的原非水溶性抗原。水相溶液所含有的水溶性抗原的浓度或原非水溶性抗原的浓度,也即第一预定浓度要求蛋白质多肽浓度含量大于0.01ng/mL,能负载足够癌细胞全细胞抗原以激活相关细胞。免疫增强佐剂在初始水相中的浓度为大于0.01ng/mL。In some embodiments, the aqueous solution contains each component in the tumor tissue lysate and an immune-enhancing adjuvant; each component in the tumor tissue lysate is water-soluble antigen or dissolved in urea or guanidine hydrochloride, respectively, during preparation The original water-insoluble antigen in the isolytic agent. The concentration of the water-soluble antigen contained in the aqueous solution or the concentration of the original water-insoluble antigen, that is, the first predetermined concentration requires the protein and polypeptide concentration to be greater than 0.01 ng/mL, which can load enough cancer cell whole cell antigens to activate relevant cells. The concentration of the immunopotentiating adjuvant in the initial aqueous phase was greater than 0.01 ng/mL.
在一些实施例中,制备粒子原材料为PLGA,有机溶剂选用二氯甲烷。另外,在一些实施例中,制备粒子原材料的第二预定浓度的范围为0.5mg/mL-5000mg/mL,优选为100mg/mL。In some embodiments, the raw material for preparing the particles is PLGA, and the organic solvent is dichloromethane. Additionally, in some embodiments, the second predetermined concentration of the preparation particle raw material ranges from 0.5 mg/mL to 5000 mg/mL, preferably 100 mg/mL.
在本发明中,之所以选择PLGA或修饰的PLGA,是由于该材料为生物可降解材料且已被FDA批准用作药物敷料。研究表明PLGA具有一定的免疫调节功能,因而适合作为纳米粒子或微米粒子制备时的辅料。在实际应用中可根据实际情况选择合适的材料。In the present invention, PLGA or modified PLGA is chosen because the material is biodegradable and has been approved by the FDA for use as a drug dressing. Studies have shown that PLGA has certain immunomodulatory functions, so it is suitable as an excipient for the preparation of nanoparticles or microparticles. In practical applications, appropriate materials can be selected according to the actual situation.
实际中,有机相的第二预定体积根据其和水相的第一预定体积的比例进行设定,在本发明中,水相的第一预定体积和有机相的第二预定体积之比的范围为1:1.1-1:5000,优选地为1:10。在具体实施过程中可根据需要对第一预定体积、第二预定体积和第一预定体积与第二预定体积之比进行调整以调整制备的纳米粒或微米粒的尺寸大小。In practice, the second predetermined volume of the organic phase is set according to its ratio to the first predetermined volume of the water phase. In the present invention, the range of the ratio of the first predetermined volume of the water phase to the second predetermined volume of the organic phase is 1:1.1-1:5000, preferably 1:10. In the specific implementation process, the first predetermined volume, the second predetermined volume and the ratio of the first predetermined volume to the second predetermined volume can be adjusted as required to adjust the size of the prepared nanoparticles or microparticles.
优选地,水相溶液为裂解物组分溶液时,其中蛋白质和多肽的浓度大于1ng/mL,优选1mg/mL~100mg/mL;水相溶液为裂解物组分/免疫佐剂溶液时,其中蛋白质和多肽的浓度大于1ng/mL,优选1mg/mL~100mg/mL,免疫佐剂的浓度大于0.01ng/mL,优选0.01mg/mL~20mg/mL。有机相溶液中,溶剂为DMSO、乙腈、乙醇、氯仿、甲醇、DMF、异丙醇、二氯甲烷、丙醇、乙酸乙酯等,优选二氯甲烷;有机相的浓度为0.5mg/mL~5000mg/mL,优选为100mg/mL。Preferably, when the aqueous phase solution is a lysate component solution, the concentration of proteins and polypeptides is greater than 1 ng/mL, preferably 1 mg/mL to 100 mg/mL; when the aqueous phase solution is a lysate component/immune adjuvant solution, wherein The concentration of protein and polypeptide is greater than 1 ng/mL, preferably 1 mg/mL to 100 mg/mL, and the concentration of immune adjuvant is greater than 0.01 ng/mL, preferably 0.01 mg/mL to 20 mg/mL. In the organic phase solution, the solvent is DMSO, acetonitrile, ethanol, chloroform, methanol, DMF, isopropanol, dichloromethane, propanol, ethyl acetate, etc., preferably dichloromethane; the concentration of the organic phase is 0.5mg/mL~ 5000 mg/mL, preferably 100 mg/mL.
步骤2,将步骤1得到的混合液进行大于2秒的超声处理或大于1分钟的搅拌或均质处理或微流控处理。优选地,搅拌为机械搅拌或者磁力搅拌时,搅拌速度大于50rpm,搅拌时间大于1分钟,比如搅拌速度为50rpm~1500rpm,搅拌时间为0.1小时~24小时;超声处理时,超声功率大于5W,时间大于0.1秒,比如2~200秒;均质处理时使用高压/超高压均质机或高剪切均质机,使用高压/超高压均质机时压力大于5psi,比如20psi~100psi,使用高剪切均质机时转速大于100rpm,比如1000rpm~5000rpm;使用微流控处理流速大于0.01mL/min,比如0.1mL/min-100mL/min。超声或者搅拌或者均质处理或者微流控处理进行纳米化和/或微米化,超声时间长短或搅拌速度或均质处理压力及时间能控制制备的微纳粒子大小,过大或过小都会带来粒径大小的变化。In
步骤3,将步骤2处理后得到的混合物加入第三预定体积的含有第三预定浓度乳化剂的水溶液中并进行大于2秒的超声处理或大于1分钟的搅拌或进行均质处理或微流控处理。该步骤将步骤2得到的混合物加入到乳化剂水溶液中继续超声或搅拌纳米化或微米化。在本发明中,超声时间大于0.1秒,比如2~200秒,搅拌速度大于50rpm,比如50rpm~500rpm,搅拌时间大于1分钟,比如60~6000秒。优选地,搅拌为机械搅拌或者磁力搅拌时,搅拌速度大于50rpm,搅拌时间大于1分钟,比如搅拌速度为50rpm~1500rpm,搅拌时间为0.5小时~5小时;超声处理时,超声功率为50W~500W,时间大于0.1秒,比如2~200秒;均质处理时使用高压/超高压均质机或高剪切均质机,使用高压/超高压均质机时压力大于20psi,比如20psi~100psi,使用高剪切均质机时转速大于1000rpm,比如1000rpm~5000rpm;使用微流控处理流速大于0.01mL/min,比如0.1mL/min-100mL/min。超声或者搅拌或者均质处理或者微流控处理进行纳米化或微米化,超声时间长短或搅拌速度或均质处理压力及时间能控制制备的纳米或微米粒子大小,过大或过小都会带来粒径大小的变化。
在一些实施例中,乳化剂水溶液为聚乙烯醇(PVA)水溶液,第三预定体积为5mL,第三预定浓度为20mg/mL。第三预定体积根据其与第二预定体积的比例进行调整。在本发明中,第二预定体积与第三预定体积之的范围为1:1.1-1:1000进行设定,优选地可以为2:5。在具体实施过程中为了控制纳米粒子或微米粒子的尺寸,可以对第二预定体积和第三预定体积之比进行调整。同样地,本步骤的超声时间或搅拌时间、乳化剂水溶液的体积以及浓度的取值根据,均为了得到尺寸大小合适的纳米粒或微米粒。In some embodiments, the aqueous emulsifier solution is an aqueous polyvinyl alcohol (PVA) solution, the third predetermined volume is 5 mL, and the third predetermined concentration is 20 mg/mL. The third predetermined volume is adjusted according to its ratio to the second predetermined volume. In the present invention, the range between the second predetermined volume and the third predetermined volume is set to be 1:1.1-1:1000, preferably 2:5. In the specific implementation process, in order to control the size of the nano-particles or micro-particles, the ratio of the second predetermined volume to the third predetermined volume can be adjusted. Similarly, the ultrasonic time or stirring time in this step, the volume and concentration of the emulsifier aqueous solution are based on the values to obtain nanoparticles or microparticles of suitable size.
步骤4,将步骤3处理后得到的液体加入第四预定体积的第四预定浓度的乳化剂水溶液中,并进行搅拌直至满足预定搅拌条件。In
本步骤中,乳化剂水溶液为PVA溶液或其他溶液。In this step, the aqueous emulsifier solution is a PVA solution or other solutions.
第四预定浓度为5mg/mL,第四预定浓度的选择,以得到尺寸大小合适的纳米粒或微米粒为依据。第四预定体积的选择依据第三预定体积与第四预定体积之比决定。在本发明中,第三预定体积与第三预定体积之比为范围为1:1.5-1:2000,优选地为1:10。在具体实施过程中为了控制纳米粒子或微米粒子的尺寸可以对第三预定体积和第四预定体积之比进行调整。The fourth predetermined concentration is 5 mg/mL, and the selection of the fourth predetermined concentration is based on obtaining nanoparticles or microparticles of suitable size. The selection of the fourth predetermined volume is determined according to the ratio of the third predetermined volume to the fourth predetermined volume. In the present invention, the ratio of the third predetermined volume to the third predetermined volume is in the range of 1:1.5-1:2000, preferably 1:10. In the specific implementation process, the ratio of the third predetermined volume to the fourth predetermined volume may be adjusted in order to control the size of the nano-particles or micro-particles.
在本发明中,本步骤的预定搅拌条件为直至有机溶剂挥发完成,也即步骤1中的二氯甲烷挥发完成。In the present invention, the predetermined stirring condition in this step is until the volatilization of the organic solvent is completed, that is, the volatilization of the dichloromethane in
步骤5,将步骤4处理满足预定搅拌条件的混合液在以大于100RPM的转速进行大于1分钟的离心后,去除上清液,并将剩下的沉淀物重新混悬于第五预定体积的第五预定浓度的含有冻干保护剂的水溶液中或者第六预定体积的PBS(或生理盐水)中。Step 5: After centrifuging the mixture that meets the predetermined stirring conditions in
在本发明一些实施方案中,步骤5所得沉淀重新混悬于第六预定体积的PBS(或生理盐水)中时不需要冻干,可直接进行后续纳米粒子或微米粒子表面吸附癌细胞裂解物的相关实验。In some embodiments of the present invention, when the pellet obtained in step 5 is resuspended in a sixth predetermined volume of PBS (or physiological saline), it does not need to be lyophilized, and the subsequent adsorption of cancer cell lysate on the surface of nanoparticles or microparticles can be carried out directly. related experiments.
在本发明一些实施方案中,步骤5所得沉淀重新混悬于含有冻干保护剂的水溶液中时需进行冷冻干燥,再冷冻干燥以后再进行后续纳米粒子或微米粒子表面吸附癌细胞裂解物的相关实验。In some embodiments of the present invention, when the precipitate obtained in step 5 is resuspended in an aqueous solution containing a lyoprotectant, it needs to be freeze-dried, and after freeze-drying, the subsequent adsorption of cancer cell lysates on the surface of nanoparticles or microparticles is carried out. experiment.
在本发明中,该步骤的冻干保护剂的第五预定浓度为质量百分比4%,之所以如此设定,是为了在后续进行冷冻干燥中不影响冻干效果。In the present invention, the fifth predetermined concentration of the freeze-drying protective agent in this step is 4% by mass, and the reason for this setting is to not affect the freeze-drying effect in the subsequent freeze-drying.
步骤6,将步骤5得到的含有冻干保护剂的混悬液进行冷冻干燥处理后,将冻干物质备用。
步骤7,将第六预定体积的步骤5中得到的重悬于PBS(或生理盐水)中的含纳米粒的混悬液或者采用第六预定体积的PBS(或生理盐水)重悬步骤6得到的冷冻干燥后的含有纳米粒或微米粒和冻干保护剂的冻干物质直接使用;或者上述样品与第七预定体积的水溶性抗原或者溶解的原非水溶性抗原混合后使用。Step 7, resuspend the nanoparticle-containing suspension obtained in step 5 of the sixth predetermined volume in PBS (or physiological saline) or use the sixth predetermined volume of PBS (or physiological saline) to resuspend the suspension obtained in
在本发明中,第六预定体积与第七预定体积的体积比为1:10000到10000:1,优先体积比为1:100到100:1,最优体积比为1:30到30:1。In the present invention, the volume ratio of the sixth predetermined volume to the seventh predetermined volume is 1:10000 to 10000:1, the preferred volume ratio is 1:100 to 100:1, and the optimal volume ratio is 1:30 to 30:1 .
在一些实施例中,所述重悬的纳米粒子混悬液体积为10mL时,含有癌细胞裂解物或含有肿瘤组织裂解物中的水溶性抗原或者溶解的原非水溶性抗原的体积与为1mL。在实际使用时可将二者体积和比例根据需要进行调整。In some embodiments, when the volume of the resuspended nanoparticle suspension is 10 mL, the volume of the resuspended nanoparticle suspension containing the cancer cell lysate or the water-soluble antigen contained in the tumor tissue lysate or the dissolved original water-insoluble antigen is 1 mL. . In actual use, the volume and ratio of the two can be adjusted as needed.
步骤8,将抗原提呈细胞与上述制备的纳米粒子和/或微米粒子共孵育一定时间。制备纳米粒子和/微米粒子的肿瘤组织和/或癌细胞与抗原提呈细胞可以来自于自体或者同种异体。Step 8, co-incubating the antigen-presenting cells with the above-prepared nanoparticles and/or microparticles for a certain period of time. Tumor tissue and/or cancer cells and antigen-presenting cells from which nanoparticles and/or microparticles are prepared can be derived from autologous or allogeneic sources.
步骤9,收集共孵育后的抗原提呈细胞,可以进行洗涤或不经洗涤后进行机械破坏或者添加化学物质使细胞结构破坏但是仍保存细胞膜组分,所使用的机械破坏或添加化学物质的方法包括但不限于进行低于500W的超声、高压均质化、搅拌、溶胀、皱缩、高剪切力破坏、小孔径挤压破坏、挤出、添加含有化学物质的低渗透压PBS或葡萄糖溶液或者盐溶液中的一种或多种。Step 9, collect the antigen-presenting cells after co-incubation, which can be washed or not washed and then mechanically destroyed or added with chemical substances to destroy the cell structure but still preserve the cell membrane components, the method of mechanical destruction or chemical addition used Including but not limited to sonication below 500W, high pressure homogenization, agitation, swelling, shrinking, high shear failure, small pore size extrusion failure, extrusion, addition of low osmotic pressure PBS or glucose solution containing chemicals Or one or more of the salt solutions.
步骤10,将机械破坏或者添加特定化学物质的样品进行离心、和/或通过特定孔径的滤膜过滤、挤出、和/或与负载全细胞组分的纳米粒子或微米粒子共作用后制备得到纳米疫苗或微米疫苗。机械破坏或者添加特定化学物质的样品与负载全细胞组分的纳米粒子或微米粒子共作用的方式包括但不限于共孵育、超声、挤出、机械搅拌、高压均质化、挤压等可以将膜组分负载到纳米粒子或微米粒子表面的方式。
在另一些实施方案中,制备负载抗原的纳米粒子或微米粒子的具体制备方法如下:In other embodiments, the specific preparation method for preparing the antigen-loaded nanoparticles or microparticles is as follows:
步骤1~4同上。
步骤5,将步骤4处理满足预定搅拌条件的混合液在以大于100RPM的转速进行大于1分钟的离心后,去除上清液,并将剩下的沉淀物重新混悬于第五预定体积的第五预定浓度的含有癌细胞全细胞抗原中水溶性和/或非水溶性抗原的溶液中,或者将剩下的沉淀物重新混悬于第五预定体积的第五预定浓度的含有癌细胞全细胞抗原中水溶性和/或非水溶性抗原与佐剂混合的溶液中。Step 5: After centrifuging the mixture that meets the predetermined stirring conditions in
步骤6,将步骤5处理满足预定搅拌条件的混合液在以大于100RPM的转速进行大于1分钟的离心后,去除上清液,并将剩下的沉淀物重新混悬于第六预定体积的固化处理试剂或矿化处理试剂,作用一定时间后离心洗涤,然后加入第七预定提交的含有带正电或者带负电的物质并作用一定时间。Step 6: After centrifuging the mixture that meets the predetermined stirring conditions in step 5 at a speed of more than 100 RPM for more than 1 minute, remove the supernatant, and resuspend the remaining precipitate in a sixth predetermined volume of solidification. The treatment reagent or the mineralization treatment reagent, after acting for a certain period of time, is centrifuged and washed, and then the seventh predetermined submission containing the positively charged or negatively charged substance is added and acted for a certain period of time.
在本发明一些实施方案中,步骤6所得沉淀重新混悬于第七预定体积的带电物质后可不需要冻干,可直接进行后续纳米粒子或微米粒子表面负载癌细胞/组织裂解物的相关实验。In some embodiments of the present invention, after the pellet obtained in
在本发明一些实施方案中,步骤6所得沉淀重新混悬于含有干燥保护剂的水溶液中后进行室温真空干燥或者冷冻真空干燥,在干燥以后再进行后续纳米粒子或微米粒子表面吸附癌细胞裂解物的相关实验。In some embodiments of the present invention, the precipitate obtained in
在本发明中,所述冻干保护剂选用海藻糖(Trehalose),或者甘露醇与蔗糖的混合溶液。在本发明中,该步骤的干燥保护剂的浓度为质量百分比4%,之所以如此设定,是为了在后续进行干燥中不影响干燥效果。In the present invention, Trehalose or a mixed solution of mannitol and sucrose is selected as the freeze-drying protective agent. In the present invention, the concentration of the drying protective agent in this step is 4% by mass, which is set so as not to affect the drying effect in the subsequent drying.
步骤7,将步骤6得到的含有干燥保护剂的混悬液进行干燥处理后,将干燥后的物质备用。In step 7, after drying the suspension containing the drying protective agent obtained in
步骤8,将第八预定体积的步骤6中得到的重悬于PBS(或生理盐水)中的含纳米粒的混悬液或者采用第八预定体积的PBS(或生理盐水)重悬步骤7得到的干燥后的含有纳米粒或微米粒和干燥保护剂的干燥后物质直接使用;或者与第九预定体积的水溶性抗原或者非水溶性抗原混合后使用。Step 8, resuspend the nanoparticle-containing suspension obtained in
在本发明中,步骤5-步骤8的修饰和抗原负载步骤可重复多次以提高抗原的负载量。而且在添加带正电或带负电的物质时可以多次添加带同种电荷的或者也可以交替添加带不同电荷的物质。In the present invention, the modification and antigen loading steps of steps 5 to 8 can be repeated multiple times to increase the antigen loading. Moreover, when adding positively charged or negatively charged substances, substances with the same charge can be added multiple times, or substances with different charges can be added alternately.
在一些实施例中,所述重悬的纳米粒子混悬液体积为10mL时,含有癌细胞裂解物或含有肿瘤组织裂解物中的水溶性抗原或者原非水溶性抗原的体积与为0.1-100mL。在实际使用时可将二者体积和比例根据需要进行调整。In some embodiments, when the volume of the resuspended nanoparticle suspension is 10 mL, the volume of the resuspended nanoparticle suspension containing the cancer cell lysate or containing the water-soluble antigen in the tumor tissue lysate or the original water-insoluble antigen is 0.1-100 mL. . In actual use, the volume and ratio of the two can be adjusted as needed.
步骤9,将抗原提呈细胞与上述制备的纳米粒子和/或微米粒子共孵育一定时间。制备纳米粒子和/微米粒子的肿瘤组织和/或癌细胞与抗原提呈细胞可以来自于自体或者同种异体。Step 9, co-incubating the antigen-presenting cells with the above-prepared nanoparticles and/or microparticles for a certain period of time. Tumor tissue and/or cancer cells and antigen-presenting cells from which nanoparticles and/or microparticles are prepared can be derived from autologous or allogeneic sources.
步骤10,收集共孵育后的抗原提呈细胞,可以进行洗涤或不经洗涤后进行机械破坏或者添加化学物质使细胞结构破坏但是仍保存细胞膜组分,所使用的机械破坏或添加化学物质的方法包括但不限于进行低于500W的超声、高压均质化、搅拌、挤出、溶胀、皱缩、高剪切力破坏、小孔径挤压破坏、添加含有化学物质的低渗透压PBS或葡萄糖溶液或者盐溶液中的一种或多种。Step 10: Collect the antigen-presenting cells after co-incubation, which can be washed or not washed and then mechanically destroyed or added with chemical substances to destroy the cell structure but still preserve the cell membrane components, the method of mechanical destruction or chemical addition used Including but not limited to sonication below 500W, high pressure homogenization, stirring, extrusion, swelling, shrinking, high shear failure, small pore size extrusion failure, addition of low osmotic pressure PBS or glucose solution containing chemicals Or one or more of the salt solutions.
步骤11,将机械破坏或者添加特定化学物质的样品进行离心、和/或通过特定孔径的滤膜过滤、和/或与负载全细胞组分的纳米粒子或微米粒子共作用后制备得到纳米疫苗或微米疫苗。机械破坏或者添加特定化学物质的样品与负载全细胞组分的纳米粒子或微米粒子共作用的方式包括但不限于共孵育、超声、机械搅拌、挤出、高压均质化、挤压等可以将膜组分负载到纳米粒子或微米粒子表面的方式。Step 11, centrifuging the mechanically damaged or added sample with specific chemical substances, and/or filtering through a filter membrane with a specific pore size, and/or co-acting with nanoparticles or microparticles loaded with whole cell components to prepare a nanovaccine or a nanovaccine. Micron vaccine. Means of mechanical disruption or addition of specific chemical substances to the co-action of the sample with the whole-cell component-loaded nanoparticles or microparticles include, but are not limited to, co-incubation, sonication, mechanical stirring, extrusion, high pressure homogenization, extrusion, etc. The manner in which membrane components are loaded onto the surface of nanoparticles or microparticles.
实施例1DC来源的纳米疫苗用于黑色素瘤的预防Example 1DC-derived nanovaccine for the prevention of melanoma
本实施例以小鼠黑色素瘤为癌症模型来说明如何使用DC来源的纳米疫苗用于预防黑色素瘤。本实施例中,裂解B16F10黑色素瘤肿瘤组织以制备肿瘤组织的水溶性抗原和非水溶性抗原,然后,以有机高分子材料PLGA为纳米粒骨架材料,以Polyinosinic-polycytidylic acid(poly(I:C))为免疫佐剂采用溶剂挥发法制备负载有肿瘤组织的水溶性抗原和非水溶性抗原的纳米粒子系统,然后使用纳米粒子激活DC细胞,再使用DC细胞制备纳米疫苗用于预防和治疗癌症。This example uses mouse melanoma as a cancer model to illustrate how to use DC-derived nanovaccine for the prevention of melanoma. In this example, the B16F10 melanoma tumor tissue was lysed to prepare water-soluble antigens and water-insoluble antigens of the tumor tissue. Then, the organic polymer material PLGA was used as the nanoparticle skeleton material, and Polyinosinic-polycytidylic acid (poly(I:C )) using solvent volatilization method for immune adjuvant to prepare nanoparticle system loaded with water-soluble antigens and water-insoluble antigens of tumor tissue, then using nanoparticles to activate DC cells, and then using DC cells to prepare nano-vaccine for cancer prevention and treatment .
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
在每只C57BL/6小鼠背部皮下接种1.5×105个B16F10细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,通过细胞过滤网加入适量超纯水并反复冻融5次,并伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性抗原;在所得沉淀部分中加入8M尿素溶解沉淀部分即可将不溶于纯水的非水溶性抗原转化为在8M尿素水溶液中可溶。将水溶性抗原和非水溶性抗原按质量比1:1混合,即为制备纳米粒子的抗原原料来源。1.5×10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse, the mice were sacrificed when the tumors grew to a volume of about 1000 mm 3 , and tumor tissues were excised. The tumor tissue was cut into pieces and ground, and an appropriate amount of ultrapure water was added through a cell strainer and freeze-thawed 5 times, accompanied by ultrasound to destroy the lysed cells. After the cells were lysed, the lysate was centrifuged at 5000g for 5 minutes, and the supernatant was taken to be the water-soluble antigen soluble in pure water; adding 8M urea to the obtained precipitate to dissolve the precipitated part to make it insoluble in pure water. The water-insoluble antigen was converted to soluble in 8 M urea in water. Mixing the water-soluble antigen and the water-insoluble antigen in a mass ratio of 1:1 is the source of the antigen raw material for preparing the nanoparticles.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒子采用溶剂挥发法中的复乳法制备。所采用的纳米粒子制备材料PLGA分子量为24KDa-38KDa,所采用的免疫佐剂为poly(I:C)且poly(I:C)只包载于纳米粒子内。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载全细胞裂解物组分和佐剂,在内部负载细胞裂解组分和佐剂后,将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h。该纳米粒子平均粒径为250nm左右,纳米粒子表面电位为-3mV左右;每1mg PLGA纳米粒子约负载100μg蛋白质或多肽组分,每1mgPLGA纳米粒所使用的poly(I:C)免疫佐剂为0.02mg。空白纳米制备材料和制备方法相同,粒粒径为240nm左右,采用等量佐剂代替全细胞组分。In this example, the nanoparticles were prepared by the double emulsion method in the solvent evaporation method. The used nanoparticle preparation material PLGA has a molecular weight of 24KDa-38KDa, and the used immune adjuvant is poly(I:C) and poly(I:C) is only contained in the nanoparticle. The preparation method was as described above. In the preparation process, the whole cell lysate component and adjuvant were first loaded into the nanoparticles by double emulsion method. After the cell lysate component and the adjuvant were loaded inside, 100 mg of nanoparticles were centrifuged at 10,000 g for 20 minutes. , and resuspended in 10 mL of ultrapure water containing 4% trehalose and freeze-dried for 48 h. The average particle size of the nanoparticles is about 250nm, and the surface potential of the nanoparticles is about -3mV; each 1mg PLGA nanoparticles is loaded with about 100μg protein or polypeptide components, and the poly(I:C) immune adjuvant used per 1mg PLGA nanoparticles is 0.02mg. The blank nanometer preparation material and preparation method are the same, the particle size is about 240nm, and the same amount of adjuvant is used to replace the whole cell component.
(3)骨髓来源的树突状细胞(BMDC)的制备(3) Preparation of bone marrow-derived dendritic cells (BMDC)
本实施例以从小鼠骨髓细胞制备树突状细胞为例来说明如何制备BMDC。首先,取1只6-8周龄C57小鼠颈椎脱臼处死,手术取出后腿的胫骨和股骨放入PBS中,用剪刀和镊子将骨周围的肌肉组织剔除干净。用剪刀剪去骨头两端,再用注射器抽取PBS溶液,针头分别从骨头两端插入骨髓腔,反复冲洗骨髓到培养皿中。收集骨髓溶液,400g离心3min后加入1mL红细胞裂解液裂红。加入3mL RPMI 1640(10%FBS)培养基终止裂解,400g离心3min,弃上清。将细胞放置10mm培养皿中培养,使用RPMI 1640(10%FBS)培养基,同时加入重组小鼠GM-CSF(20ng/mL),37度,5%CO2培养7天。第3天轻轻摇晃培养瓶,补充同样体积含有GM-CSF(20ng/mL)RPMI 1640(10%FBS)培养基。第6天,对培养基进行半量换液处理。第7天,收集少量悬浮及半贴壁细胞,通过流式检测,当CD86+CD80+细胞在CD11c+细胞中的比例为15-20%之间,诱导培养的BMDC即可被用来做下一步实验。This example uses the preparation of dendritic cells from mouse bone marrow cells as an example to illustrate how to prepare BMDCs. First, a 6-8-week-old C57 mouse was sacrificed by cervical dislocation. The tibia and femur of the hind leg were surgically removed and placed in PBS. The muscle tissue around the bone was removed with scissors and forceps. Cut off both ends of the bone with scissors, and then use a syringe to extract the PBS solution. The needles are inserted into the bone marrow cavity from both ends of the bone, and the bone marrow is repeatedly washed into the Petri dish. The bone marrow solution was collected, centrifuged at 400 g for 3 min, and then 1 mL of erythrocyte lysate was added. 3 mL of RPMI 1640 (10% FBS) medium was added to terminate the lysis, centrifuged at 400 g for 3 min, and the supernatant was discarded. The cells were placed in a 10 mm petri dish and cultured in RPMI 1640 (10% FBS) medium with the addition of recombinant mouse GM-CSF (20 ng/mL) at 37 degrees, 5% CO 2 for 7 days. On
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载来源于肿瘤组织的癌细胞全细胞组分的纳米粒子(500μg)或空白纳米粒(500μg)+游离裂解液与BMDC(1000万个)在15mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有细胞因子组合1:粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(1000U/mL)或者含有细胞因子组分2:GM-CSF,(1000U/mL)、IL-4(100U/mL)、肿瘤坏死因子α(TNF-α,200U/mL)。Nanoparticles (500 μg) or blank nanoparticles (500 μg) + free lysate loaded with whole cell fractions of cancer cells derived from tumor tissue were incubated with BMDC (10 million) in 15 mL of RPMI1640 complete medium for 48 hours (37°C). , 5% CO 2 ); the incubation system contains cytokine combination 1: granulocyte-macrophage colony stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL) mL), IL-12 (1000U/mL), or with cytokine components 2: GM-CSF, (1000U/mL), IL-4 (100U/mL), tumor necrosis factor alpha (TNF-α, 200U/mL) ).
(5)DC来源的纳米疫苗的制备(5) Preparation of DC-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC,然后使用生理盐水洗涤细胞两遍,将细胞重悬在生理盐水中后在7.5W超声20分钟。然后将样品在2000g离心20分钟并收集上清液,将上清液在7000g离心20分钟后收集上清液,然后在15000g离心120分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗,纳米疫苗粒径为120纳米。The incubated DCs were collected by centrifugation at 400g for 5 minutes, then the cells were washed twice with saline, resuspended in saline and sonicated at 7.5W for 20 minutes. The samples were then centrifuged at 2000g for 20 minutes and the supernatant was collected, the supernatant was centrifuged at 7000g for 20 minutes and the supernatant was collected and then centrifuged at 15000g for 120 minutes. The supernatant was discarded to collect the pellet and the pellet was placed in PBS The nano-vaccine was obtained after resuspending, and the particle size of the nano-vaccine was 120 nm.
(6)纳米疫苗用于癌症的预防(6) Nano vaccines for cancer prevention
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在小鼠接种癌细胞之前第-42天、第-35天、第-28天、第-21天、第-14天和第-7天分别给每只小鼠皮下注射纳米疫苗500μg。在第0天,给每只小鼠背部右下方皮下接种1.5×105个B16F10细胞。监测小鼠肿瘤生长速度和小鼠生存期。在实验中,从第3天开始每3天记录一次小鼠肿瘤体积的大小。肿瘤体积采用公式v=0.52×a×b2计算,其中v为肿瘤体积,a为肿瘤长度,b为肿瘤宽度。出于动物实验伦理,在小鼠生存期试验中当小鼠肿瘤体积超过2000mm3即视为小鼠死亡并将小鼠安乐死。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. Each mouse was subcutaneously injected with 500 μg of nanovaccine on day -42, day -35, day -28, day -21, day -14 and day -7, respectively, before the mice were inoculated with cancer cells. On
(7)实验结果(7) Experimental results
如图2所示,PBS对照组的小鼠其肿瘤生长速度很快,生存期很短。接收空白纳米粒+游离裂解液激活的DC制备的纳米疫苗处理的小鼠其肿瘤生长速度变慢。接受负载癌细胞全细胞裂解物的纳米粒子激活的DC制备的纳米疫苗处理的小鼠其肿瘤生长速度明显比上述两组更慢,生存期最长。其中,DC细胞激活过程中加入细胞因子组合1的效果好于细胞因子组合2。综上所述,本发明所述纳米疫苗对黑色素瘤具有良好的预防效果。As shown in Figure 2, mice in the PBS control group had fast tumor growth and short survival. Mice treated with nanovaccine prepared from blank nanoparticles + free lysate-activated DC had slower tumor growth. Mice treated with the nanovaccine prepared from nanoparticle-activated DCs loaded with whole-cell lysates of cancer cells had significantly slower tumor growth and longest survival than the above two groups. Among them, the effect of adding
实施例2基于DC的癌症疫苗用于黑色素瘤的预防Example 2 DC-based cancer vaccines for melanoma prevention
本实施例以小鼠黑色素瘤为癌症模型来说明如何使用纳米粒子辅助激活DC后,将DC细胞制备成癌症疫苗,用于预防黑色素瘤。本实施例中,裂解B16F10黑色素瘤肿瘤组织以制备肿瘤组织的水溶性抗原和非水溶性抗原,然后,以有PLGA为纳米粒骨架材料,以同为Toll样受体激动剂的poly(I:C)、CpG2216和CpG2395为混合免疫佐剂采用溶剂挥发法制备负载有肿瘤组织的水溶性抗原和非水溶性抗原的纳米粒子系统,然后使用纳米粒子激活DC,将DC经过适当处理后制备成源于DC的纳米疫苗注射到体内预防黑色素瘤。This example uses mouse melanoma as a cancer model to illustrate how to use nanoparticles to assist in activating DCs, and then prepare DCs into cancer vaccines for the prevention of melanoma. In this example, the B16F10 melanoma tumor tissue was split to prepare water-soluble antigens and water-insoluble antigens of the tumor tissue, then, PLGA was used as the nanoparticle skeleton material, and poly(I: C), CpG2216 and CpG2395 are mixed immune adjuvants to prepare nanoparticle systems loaded with water-soluble antigens and non-water-soluble antigens of tumor tissue by solvent evaporation method, and then use nanoparticles to activate DCs, and DCs are prepared into sources after appropriate treatment In vivo prevention of melanoma by injection of nanovaccine on DC.
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
在每只C57BL/6小鼠背部皮下接种1.5×105个B16F10细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,通过细胞过滤网加入适量纯水并反复冻融5次,并可伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性抗原;在所得沉淀部分中加入8M尿素溶解沉淀部分即可将不溶于纯水的非水溶性抗原转化为在8M尿素水溶液中可溶。以上即为制备纳米粒子系统的抗原原料来源。1.5×10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse, the mice were sacrificed when the tumors grew to a volume of about 1000 mm 3 , and tumor tissues were excised. The tumor tissue was cut into pieces and ground, and an appropriate amount of pure water was added through a cell strainer and freeze-thawed for 5 times. Ultrasound was also used to destroy the lysed cells. After the cells were lysed, the lysate was centrifuged at 5000g for 5 minutes, and the supernatant was taken to be the water-soluble antigen soluble in pure water; adding 8M urea to the obtained precipitate to dissolve the precipitated part to make it insoluble in pure water. The water-insoluble antigen was converted to soluble in 8 M urea in water. The above is the source of antigen raw materials for the preparation of nanoparticle systems.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米疫苗及作为对照的空白纳米粒采用溶剂挥发法制备。在制备时负载癌细胞全细胞抗原中水溶性抗原的纳米疫苗和负载癌细胞全细胞抗原中非水溶性抗原的纳米粒子分别制备,然后使用时一起使用。所采用的纳米粒子制备材料PLGA分子量为7Da-17KDa,所采用的免疫佐剂为poly(I:C)、CpG2216和CpG2395且佐剂包载于纳米粒子内部。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载抗原和佐剂,在内部负载抗原后,将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h。该纳米粒子平均粒径为280nm左右;每1mg PLGA纳米粒子约负载100μg蛋白质和多肽组分,每1mgPLGA纳米粒所使用的poly(I:C)、CpG2216和CpG2395免疫佐剂各0.025mg。本实施例中,采用等质量负载四种多肽新生抗原B16-M20(Tubb3,FRRKAFLHWYTGEAMDEMEFTEAESNM),B16-M24(Dag1,TAVITPPTTTTKKARVSTPKPATPSTD),B16-M46(Actn4,NHSGLVTFQAFIDVMSRETTDTDTADQ)和TRP2:180-188(SVYDFFVWL)的纳米粒子作为对照纳米粒子使用,对照纳米粒粒径为260nm左右,负载100μg多肽组分,负载等量佐剂。空白纳米粒粒径为250nm左右,只负载等量的免疫佐剂却不负载任何抗原组分。In this example, the nanovaccine and the blank nanoparticle as a control were prepared by solvent evaporation method. During preparation, the nanovaccine loaded with the water-soluble antigen in the cancer cell whole cell antigen and the nanoparticle loaded with the water-insoluble antigen in the cancer cell whole cell antigen are prepared separately, and then used together when used. The used nanoparticle preparation material PLGA has a molecular weight of 7Da-17KDa, and the used immune adjuvants are poly(I:C), CpG2216 and CpG2395, and the adjuvants are encapsulated inside the nanoparticles. The preparation method was as described above. In the preparation process, the antigen and adjuvant were first loaded into the nanoparticles by the double emulsion method. After the antigen was loaded inside, 100 mg of the nanoparticles were centrifuged at 10,000 g for 20 minutes, and 10 mL of 4% trehalose was used. Resuspend in ultrapure water and freeze-dry for 48 h. The average particle size of the nanoparticles is about 280 nm; each 1 mg of PLGA nanoparticles is loaded with about 100 μg of protein and polypeptide components, and each 1 mg of PLGA nanoparticles uses 0.025 mg of poly(I:C), CpG2216 and CpG2395 immune adjuvants. In this example, four polypeptide neoantigens B16-M20 (Tubb3, FRRKAFLHWYTGEAMDEMEFTEAESNM), B16-M24 (Dag1, TAVITPPTTTTKKARVSTPKPATPSTD), B16-M46 (Actn4, NHSGLVTFQAFIDVMSRETTDTDTADQ) and TRP2:180-188 (SVYDFFVWL) were loaded with equal mass. The nanoparticles were used as control nanoparticles. The size of the control nanoparticles was about 260 nm, loaded with 100 μg of polypeptide components, and loaded with an equal amount of adjuvant. The particle size of the blank nanoparticles is about 250 nm, and only the same amount of immune adjuvant is loaded without any antigenic components.
(3)骨髓来源的树突状细胞(BMDC)的制备(3) Preparation of bone marrow-derived dendritic cells (BMDC)
本实施例以从小鼠骨髓细胞制备树突状细胞为例来说明如何制备BMDC。首先,取1只6-8周龄C57小鼠颈椎脱臼处死,手术取出后腿的胫骨和股骨放入PBS中,用剪刀和镊子将骨周围的肌肉组织剔除干净。用剪刀剪去骨头两端,再用注射器抽取PBS溶液,针头分别从骨头两端插入骨髓腔,反复冲洗骨髓到培养皿中。收集骨髓溶液,400g离心3min后加入1mL红细胞裂解液裂红。加入3mL RPMI 1640(10%FBS)培养基终止裂解,400g离心3min,弃上清。将细胞放置10mm培养皿中培养,使用RPMI 1640(10%FBS)培养基,同时加入重组小鼠GM-CSF(20ng/mL),37度,5%CO2培养7天。第3天轻轻摇晃培养瓶,补充同样体积含有GM-CSF(20ng/mL)RPMI 1640(10%FBS)培养基。第6天,对培养基进行半量换液处理。第7天,收集少量悬浮及半贴壁细胞,通过流式检测,当CD86+CD80+细胞在CD11c+细胞中的比例为15-20%之间,诱导培养的BMDC即可被用来做下一步实验。This example uses the preparation of dendritic cells from mouse bone marrow cells as an example to illustrate how to prepare BMDCs. First, a 6-8-week-old C57 mouse was sacrificed by cervical dislocation. The tibia and femur of the hind leg were surgically removed and placed in PBS. The muscle tissue around the bone was removed with scissors and forceps. Cut off both ends of the bone with scissors, and then use a syringe to extract the PBS solution. The needles are inserted into the bone marrow cavity from both ends of the bone, and the bone marrow is repeatedly washed into the Petri dish. The bone marrow solution was collected, centrifuged at 400 g for 3 min, and then 1 mL of erythrocyte lysate was added. 3 mL of RPMI 1640 (10% FBS) medium was added to terminate the lysis, centrifuged at 400 g for 3 min, and the supernatant was discarded. The cells were placed in a 10 mm petri dish and cultured in RPMI 1640 (10% FBS) medium with the addition of recombinant mouse GM-CSF (20 ng/mL) at 37 degrees, 5% CO 2 for 7 days. On
(4)DC的激活(4) Activation of DC
将负载来源于肿瘤组织的癌细胞全细胞组分的纳米粒子(500μg,其中负载水溶性组分的纳米粒子250μg,负载非水溶性组分的纳米粒子250μg)或多肽纳米粒子(500μg)或空白纳米粒(500μg)+游离裂解液与BMDC(1000万个)在15mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO2),孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(1000U/mL)、IL-12(500U/mL)和CD86抗体(20ng/mL)。Nanoparticles (500μg, 250μg of nanoparticles loaded with water-soluble components, 250μg of nanoparticles loaded with non-water-soluble components) or polypeptide nanoparticles (500μg) or blank will be loaded with whole cell components of cancer cells derived from tumor tissue. Nanoparticles (500μg) + free lysate and BMDC (10 million) were co-incubated in 15mL RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ), and the incubation system contained granulocyte-macrophage colony-stimulating factor ( GM-CSF, 2000 U/mL), IL-2 (500 U/mL), IL-7 (1000 U/mL), IL-12 (500 U/mL) and CD86 antibody (20 ng/mL).
(5)DC来源的纳米疫苗的制备(5) Preparation of DC-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(22.5W)超声1分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,然后在16000g离心90分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后使用膜过滤器过滤样品即得纳米疫苗,纳米疫苗粒径为120纳米。Incubated DCs were collected by centrifugation at 400 g for 5 min, then cells were washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, cells were resuspended in PBS water at 4°C low power (22.5W) Sonicate for 1 minute. The samples were then centrifuged at 3000g for 15 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 8000g for 15 minutes, and then after 90 minutes at 16000g. The supernatant was discarded to collect the pellet and the pellet was placed in PBS After resuspension, a membrane filter was used to filter the sample to obtain a nano-vaccine, and the particle size of the nano-vaccine was 120 nm.
(6)纳米疫苗用于癌症的预防(6) Nano vaccines for cancer prevention
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠,在小鼠接种癌细胞前第-35天、第-28天、第-21天、第-14天和第-7天每只小鼠分别接种纳米疫苗500μg。在第0天,给每只受体小鼠背部右下方皮下接种1.5×105个B16F10细胞。监测小鼠肿瘤生长速度和小鼠生存期。在实验中,从第3天开始每3天记录一次小鼠肿瘤体积的大小。肿瘤体积采用公式v=0.52×a×b2计算,其中v为肿瘤体积,a为肿瘤长度,b为肿瘤宽度。出于动物实验伦理,在小鼠生存期试验中当小鼠肿瘤体积超过2000mm3即视为小鼠死亡并将小鼠安乐死。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice, on the -35th, -28th, -21st, -14th and -7 days each mouse was vaccinated with 500 μg of nanovaccine. On
(7)纳米疫苗激活癌细胞特异性T细胞的分析(7) Analysis of nano-vaccine activation of cancer cell-specific T cells
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在第0天、第7天、第14天、第28天和第42天分别每只小鼠皮下注射100μg纳米疫苗或PBS。在第45天处死小鼠,摘取小鼠脾脏并制备脾细胞单细胞悬液,使用磁珠分选法从小鼠脾细胞中分选出B细胞和T细胞。将100μg负载全细胞组分的纳米粒子、500万个B细胞和100万个T细胞在5mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO2)。然后收集孵育后细胞并标记活死细胞染料、CD3抗体、CD8抗体、CD4抗体和IFN-γ抗体,尔后流式细胞术分析T细胞中IFN-γ+T细胞所占比例。纳米粒子所负载的癌细胞全细胞抗原在被抗原提呈细胞B细胞吞噬后可被降解成抗原表位被提呈到抗原提呈细胞表面,可以识别癌细胞全细胞抗原的特异性T细胞即可以识别癌细胞全细胞抗原表位后被激活并分泌杀伤性细胞因子。IFN-γ是抗原特异性T细胞识别抗原后被激活所分泌的最主要的细胞因子。使用流式细胞术分析的IFN-γ+T细胞即为可以识别和杀伤癌细胞的癌细胞特异性T细胞。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. Each mouse was injected subcutaneously with 100 μg of nanovaccine or PBS on
(8)实验结果(8) Experimental results
如图3中a和b所示,接收PBS对照组和空白纳米粒对照组小鼠肿瘤生长速度都很快,小鼠生存期很短。与上述两组对照组相比,使用纳米粒激活的DC所制备的纳米疫苗处理的小鼠体内的肿瘤生长速度明显变慢,而且部分小鼠肿瘤消失痊愈。而且,负载癌细胞全细胞抗原的纳米粒子激活的DC制备的纳米疫苗效果优于负载四种抗原多肽的纳米粒子激活的DC制备的纳米疫苗。这说明负载四种新生抗原多肽纳米粒子激活的DC所处理和提呈的抗原表位种类,因而其制备的纳米疫苗所能激活的癌细胞特异性T抗原提呈细胞癌症疫苗所含有的T细胞克隆数很少,所能识别和杀灭的癌细胞也就较少。而负载癌细胞全细胞抗原的纳米粒子激活的DC处理和提呈的癌细胞抗原更广谱,因而其制备的纳米疫苗所能激活的T细胞克隆数也就更广谱,所能识别和杀灭的癌细胞也就越多,治疗或预防癌症的效果也越好。As shown in a and b in Figure 3, both the PBS control group and the blank nanoparticle control group had fast tumor growth and short survival time. Compared with the above two groups of control groups, the tumor growth rate in mice treated with nanovaccine prepared by nanoparticle-activated DC was significantly slower, and some mice tumors disappeared and recovered. Moreover, the effect of nanovaccine prepared from DCs activated by nanoparticles loaded with whole cell antigens of cancer cells is better than that prepared by DCs activated by nanoparticles loaded with four antigen polypeptides. This indicates the types of antigenic epitopes processed and presented by DCs activated by nanoparticles loaded with four neoantigen polypeptides, and thus the cancer cell-specific T antigen-presenting cells that can be activated by the prepared nanovaccine. T cells contained in cancer vaccines With fewer clones, fewer cancer cells can be identified and killed. However, the DCs activated by nanoparticles loaded with whole cell antigens of cancer cells can process and present a broader spectrum of cancer cell antigens, so the number of T cell clones that can be activated by the prepared nanovaccine is also broader, and can recognize and kill cancer cells. The more cancer cells are killed, the better the effect of treating or preventing cancer.
如图3中c和d所示,负载全细胞组分的纳米粒子激活的DC细胞制备的纳米疫苗所能激活的CD8+IFN-γ+T细胞和CD4+IFN-γ+T细胞占CD8+T细胞和CD4+T细胞的比例明显高于负载多肽的纳米粒子和空白纳米粒子+游离裂解液激活的DC细胞制备的纳米疫苗所激活的比例。由此可见,本发明所述的负载全细胞组分的纳米粒子激活的DC细胞制备的纳米疫苗可以更好的激活具有识别癌细胞和杀伤癌细胞能力的癌细胞特异性T细胞。As shown in c and d in Figure 3, the CD8+IFN-γ+T cells and CD4+IFN-γ+T cells that can be activated by the nanovaccine prepared from the DC cells activated by nanoparticles loaded with the whole cell components accounted for the proportion of CD8+ IFN-γ+ T cells. The ratios of T cells and CD4+T cells were significantly higher than those activated by the nanovaccine prepared by polypeptide-loaded nanoparticles and blank nanoparticles+DC cells activated by free lysate. It can be seen that the nano-vaccine prepared from DC cells activated by nanoparticles loaded with whole cell components according to the present invention can better activate cancer cell-specific T cells with the ability to recognize cancer cells and kill cancer cells.
实施例3Example 3
本实施例以小鼠黑色素瘤为癌症模型来说明如何使用纳米疫苗治疗癌症。本实施例中,首先裂解B16F10黑色素瘤肿瘤组织和癌细胞以制备肿瘤组织和癌细胞的水溶性抗原混合物(质量比1:1)和非水溶性抗原混合物(质量比1:1),并将水溶性抗原混合物和非水溶性抗原混合物按质量比1:1混合。然后,以PLGA为纳米粒骨架材料,以Poly(I:C)和CpG2006为佐剂制备负载裂解物组分的纳米粒子,然后将纳米粒子与T抗原提呈细胞体外共孵育一定时间,然后使用被激活的抗原提呈细胞制备纳米疫苗用于治疗癌症。This example uses mouse melanoma as a cancer model to illustrate how to use nanovaccine to treat cancer. In this example, B16F10 melanoma tumor tissue and cancer cells were firstly lysed to prepare a water-soluble antigen mixture (mass ratio 1:1) and a water-insoluble antigen mixture (mass ratio 1:1) of tumor tissue and cancer cells, and the The water-soluble antigen mixture and the water-insoluble antigen mixture were mixed in a mass ratio of 1:1. Then, nanoparticles loaded with lysate components were prepared with PLGA as nanoparticle skeleton material and Poly(I:C) and CpG2006 as adjuvants, and then the nanoparticles were co-incubated with T antigen presenting cells in vitro for a certain period of time, and then used Activated antigen-presenting cells to prepare nanovaccine for cancer treatment.
(1)肿瘤组织和癌细胞的裂解及各组分的收集(1) Lysis of tumor tissue and cancer cells and collection of components
收集肿瘤组织时先在每只C57BL/6小鼠背部皮下接种1.5×105个B16F10细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织,将肿瘤组织切块后研磨,通过细胞过滤网加入适量纯水并反复冻融5次,并可伴有超声以破坏裂解所得样品;收集培养的B16F10癌细胞系时,先离心去除培养基后使用PBS洗涤两次并离心收集癌细胞,将癌细胞在超纯水中重悬,反复冻融3次,并伴有超声破坏裂解癌细胞。待肿瘤组织或癌细胞裂解后,将裂解物以5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性抗原;在所得沉淀部分中加入8M尿素溶解沉淀部分即可将不溶于纯水的非水溶性抗原转化为在8M尿素水溶液中可溶。将肿瘤组织的水溶性抗原和癌细胞的水溶性抗原按质量比1:1混合;肿瘤组织的非水溶性抗原和癌细胞的非水溶性抗原按质量比1:1混合。将水溶性抗原混合物和非水溶性抗原混合物按质量比1:1混合,即为制备纳米粒子的抗原原料来源。When collecting tumor tissue, firstly inoculate 1.5×10 5 B16F10 cells subcutaneously on the back of each C57BL/ 6 mouse. When the tumor grows to a volume of about 1000 mm, the mice are sacrificed and the tumor tissue is excised. The tumor tissue is cut into pieces. Grind, add an appropriate amount of pure water through a cell strainer and freeze and thaw for 5 times, and sonicate to destroy the lysed samples; when collecting the cultured B16F10 cancer cell line, first centrifuge to remove the medium, then wash twice with PBS and centrifuge Cancer cells were collected, resuspended in ultrapure water, freeze-thawed 3 times, and lysed cancer cells with ultrasonic damage. After the tumor tissue or cancer cells are lysed, the lysate is centrifuged at 5000g for 5 minutes, and the supernatant is taken to be the water-soluble antigen soluble in pure water; 8M urea is added to the obtained precipitate to dissolve the precipitated part. Water-insoluble antigens that were insoluble in pure water were converted to be soluble in 8M aqueous urea. The water-soluble antigen of tumor tissue and the water-soluble antigen of cancer cells are mixed in a mass ratio of 1:1; the water-insoluble antigen of tumor tissue and the water-insoluble antigen of cancer cells are mixed in a mass ratio of 1:1. Mixing the water-soluble antigen mixture and the water-insoluble antigen mixture in a mass ratio of 1:1 is the source of antigen raw materials for preparing nanoparticles.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例中纳米粒子采用复乳法制备。所采用的纳米粒子制备材料PLGA分子量为7KDa-17KDa,所采用的免疫佐剂为poly(I:C)和CpG2006且佐剂包载于纳米粒子内。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解液组分和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h;在使用前将其用9mL PBS重悬然后加入1mL的裂解液组分(蛋白质浓度80mg/mL)并室温作用10min,得到内外都负载裂解物的纳米粒子系统。该纳米粒子平均粒径为270nm左右,表面电位为-5mV左右,每1mg PLGA纳米粒子约负载130μg蛋白质或多肽组分,每1mg PLGA纳米粒负载poly(I:C)和CpG2006免疫佐剂各0.02mg。In this example, the nanoparticles were prepared by the double emulsion method. The used nanoparticle preparation material PLGA has a molecular weight of 7KDa-17KDa, and the used immunoadjuvant is poly(I:C) and CpG2006, and the adjuvant is encapsulated in the nanoparticle. The preparation method was as described above. In the preparation process, the lysate components and adjuvants were first loaded into the nanoparticles by the double emulsion method, and then 100 mg of the nanoparticles were centrifuged at 10,000 g for 20 minutes, and 10 mL of ultrapure 4% trehalose was used. After resuspending in water, it was freeze-dried for 48 hours; before use, it was resuspended in 9 mL of PBS, then 1 mL of lysate fraction (protein concentration 80 mg/mL) was added and treated at room temperature for 10 min to obtain a nanoparticle system loaded with lysate inside and outside. The average particle size of the nanoparticles is about 270nm, the surface potential is about -5mV, each 1mg PLGA nanoparticle is loaded with about 130μg protein or polypeptide components, and each 1mg PLGA nanoparticle is loaded with poly(I:C) and CpG2006 immune adjuvant 0.02 each. mg.
(3)B细胞的分离(3) Isolation of B cells
处死C57BL/6小鼠后摘取小鼠脾脏,制备小鼠脾细胞单细胞悬液,使用磁珠分选法分离脾细胞中活细胞中(使用活死细胞染料标记死细胞以去除死细胞)的CD19+B细胞。After the C57BL/6 mice were sacrificed, the spleen of the mouse was harvested to prepare a single-cell suspension of mouse spleen cells, and the live cells in the splenocytes were separated by magnetic bead sorting (dead cells were labeled with live-dead cell dye to remove dead cells) of CD19 + B cells.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(500μg)或多肽纳米粒(500μg)与DC2.4(1000万个)和B细胞(1000万个)在15mL RPMI1640完全培养基中混合后共孵育48小时(37℃,5%CO2),孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、白蛋白(50ng/mL)、和CD80抗体(10ng/mL)。孵育完成后,将孵育后的细胞在400g离心5分钟后弃去上清液后再使用PBS洗涤两遍即得被激活的抗原提呈细胞,该被激活的混合抗原提呈细胞可作为抗原提呈细胞疫苗使用。Nanoparticles (500 μg) or polypeptide nanoparticles (500 μg) loaded with whole cell components of cancer cells were co-incubated with DC2.4 (10 million) and B cells (10 million) in 15 mL of complete RPMI1640 medium for 48 hours (37°C, 5% CO 2 ), the incubation system contains granulocyte-macrophage colony stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL) ), IL-12 (200 U/mL), albumin (50 ng/mL), and CD80 antibody (10 ng/mL). After incubation, the incubated cells were centrifuged at 400g for 5 minutes, the supernatant was discarded, and then washed twice with PBS to obtain activated antigen-presenting cells. The activated mixed antigen-presenting cells can be used as antigen-presenting cells. Cellular vaccines are used.
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集步骤(4)孵育后的DC和B细胞(2000万个,DC和B细胞各1000万个),然后使用PBS洗涤细胞三遍,将混合细胞重悬在PBS水中后在低功率(10W)超声30分钟。然后将样品在500g离心5分钟并收集上清液,将上清液依次过孔径为30μm、10μm、5μm、0.45μm、0.22μm的膜过滤后,将所得滤液样品收集后与步骤(2)制备的纳米粒子(20mg)共孵育15分钟后通过0.45μm的滤膜共挤出,然后在13000g离心25分钟后弃去上清液将沉淀使用冻干保护剂水溶液(含1%海藻糖+2%甘露醇+1%精氨酸)重悬后冷冻干燥,即得内部负载癌细胞全细胞组分同时表面负载混合抗原提呈细胞细胞膜组分的纳米疫苗,纳米疫苗粒径为280纳米。DC and B cells (20 million, 10 million each of DC and B cells) after incubation in step (4) were collected by centrifugation at 400g for 5 minutes, then washed three times with PBS, and the mixed cells were resuspended in PBS water. Sonicate at low power (10W) for 30 minutes. Then the sample was centrifuged at 500g for 5 minutes and the supernatant was collected. The supernatant was filtered through membranes with pore sizes of 30 μm, 10 μm, 5 μm, 0.45 μm and 0.22 μm in turn. The obtained filtrate samples were collected and prepared with step (2). The nanoparticles (20 mg) were co-extruded through a 0.45 μm filter after co-incubation for 15 min, and the supernatant was discarded after centrifugation at 13,000 g for 25 min. mannitol + 1% arginine) was resuspended and then freeze-dried to obtain a nano-vaccine with the whole cell components of cancer cells loaded inside and the cell membrane components of mixed antigen-presenting cells loaded on the surface. The particle size of the nano-vaccine was 280 nanometers.
(6)纳米疫苗用于癌症的治疗(6) Nano vaccines for cancer treatment
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在第0天给每只小鼠背部右下方皮下接种1.5×105个B16F10细胞。在接种黑色素瘤后第4天、第7天、第10天、第15天、第20天和第25天分别每只小鼠皮下注射100μg纳米疫苗或者皮下注射1000万个步骤(4)中激活的混合抗原提呈细胞(500万个D C+500万个B细胞)或者皮下注射100μL PBS。在实验中,从第3天开始每3天记录一次小鼠肿瘤体积的大小。肿瘤体积采用公式v=0.52×a×b2计算,其中v为肿瘤体积,a为肿瘤长度,b为肿瘤宽度。出于动物实验伦理,在小鼠生存期试验中当小鼠肿瘤体积超过2000mm3即视为小鼠死亡并将小鼠安乐死。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. 1.5 x 105 B16F10 cells were inoculated subcutaneously in the lower right back of each mouse on
(7)纳米疫苗激活癌细胞特异性T细胞的分析(7) Analysis of nano-vaccine activation of cancer cell-specific T cells
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在第0天给小鼠接种2×105个B16F10细胞,在第7天、第12天、第17天分别每只小鼠皮下注射2mg步骤(2)制备的负载癌细胞全细胞组分的纳米粒子。在第20天处死小鼠,摘取小鼠脾脏并制备脾细胞单细胞悬液,使用磁珠分选法从小鼠脾细胞中分选出B细胞和T细胞。将100μg步骤(5)制备的纳米疫苗和100万个T细胞在5mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO2);或者将1000万个步骤(4)中激活的混合抗原提呈细胞和100万个T细胞在5mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO2)。然后收集孵育后细胞并标记活死细胞染料、CD3抗体、CD8抗体、CD4抗体和IFN-γ抗体,尔后流式细胞术分析T细胞中IFN-γ+T细胞所占比例。可以识别癌细胞抗原的特异性T细胞即在识别癌细胞抗原表位后被激活并分泌杀伤性细胞因子。IFN-γ是抗原特异性T细胞识别抗原后被激活所分泌的最主要的细胞因子。使用流式细胞术分析的IFN-γ+T细胞即为可以识别和杀伤癌细胞的癌细胞特异性T细胞。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. On
(8)实验结果(8) Experimental results
如图4中a和b所示,PBS对照组小鼠的肿瘤生长速度很快,而被纳米粒子激活的抗原提呈细胞疫苗或者步骤(6)制备的纳米疫苗都可以显著减慢小鼠肿瘤生长和延长小鼠肿瘤生存期且治愈部分小鼠。而且,负载全细胞组分的纳米粒子激活的混合抗原提呈细胞制备的纳米疫苗效果好于纳米粒子激活的活的抗原提呈细胞疫苗。综上所述,本发明所述的基于抗原提呈细胞的癌症疫苗对癌症具有优异的治疗效果。As shown in a and b in Figure 4, the tumor growth rate of the mice in the PBS control group was very fast, while the antigen-presenting cell vaccine activated by nanoparticles or the nanovaccine prepared in step (6) could significantly slow down the tumor in the mice Grow and prolong tumor survival in mice and cure some mice. Moreover, the nanovaccine prepared by the nanoparticle-activated mixed antigen-presenting cells loaded with the whole-cell component is more effective than the nanoparticle-activated live antigen-presenting cell vaccine. In conclusion, the antigen-presenting cell-based cancer vaccine of the present invention has an excellent therapeutic effect on cancer.
如图4中c和d所示,步骤(6)制备的纳米疫苗所能激活的CD8+IFN-γ+T细胞和CD4+IFN-γ+T细胞占CD8+T细胞和CD4+T细胞的比例明显高于纳米粒子激活的活的混合抗原提呈细胞所能激活的比例。由此可见,与被纳米粒子激活的活的抗原提呈细胞相比,本发明所述的纳米疫苗能更好的激活具有识别癌细胞和杀伤癌细胞能力的癌细胞特异性T细胞。As shown in c and d in Figure 4, the CD8 + IFN-γ + T cells and CD4 + IFN-γ + T cells that can be activated by the nanovaccine prepared in step (6) accounted for the majority of CD8 + T cells and CD4 + T cells The ratio was significantly higher than that which could be activated by nanoparticle-activated live mixed antigen-presenting cells. It can be seen that, compared with live antigen-presenting cells activated by nanoparticles, the nanovaccine of the present invention can better activate cancer cell-specific T cells with the ability to recognize and kill cancer cells.
实施例4纳米疫苗用于黑色素瘤肺转移的预防Example 4 Nano-vaccine for the prevention of melanoma lung metastasis
本实施例以小鼠黑色素瘤肺模型来说明如何使用抗原提呈细胞来源的纳米疫苗预防癌症转移。本实施例中,首先裂解B16F10黑色素瘤肿瘤组织以制备肿瘤组织的水溶性抗原和非水溶性抗原;然后,制备负载有肿瘤组织的水溶性抗原和非水溶性抗原的纳米粒子系统。在本实施例中采用了硅化和添加带电物质的方法来增加抗原的负载量,且只进行了一轮矿化处理。本实施例中,先使用纳米粒子激活抗原提呈细胞,然后使用抗原提呈细胞制备纳米疫苗并用于预防癌症转移。This example uses a mouse melanoma lung model to illustrate how to use antigen-presenting cell-derived nanovaccine to prevent cancer metastasis. In this example, B16F10 melanoma tumor tissue was firstly lysed to prepare water-soluble antigens and water-insoluble antigens of tumor tissue; then, a nanoparticle system loaded with water-soluble antigens and water-insoluble antigens of tumor tissue was prepared. In this example, the methods of silicidation and addition of charged substances were used to increase the antigen loading, and only one round of mineralization was performed. In this example, the antigen-presenting cells are activated by using nanoparticles first, and then the nano-vaccine is prepared by using the antigen-presenting cells to prevent cancer metastasis.
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
在每只C57BL/6小鼠背部皮下接种1.5×105个B16F10细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,加入胶原酶在RPMI1640培养基中孵育30min,然后通过细胞过滤网加入适量纯水并反复冻融5次,并可伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性抗原;在所得沉淀部分中加入10%的脱氧胆酸钠溶解沉淀部分即可将不溶于纯水的非水溶性抗原转化为在10%脱氧胆酸钠水溶液中可溶,将水溶性抗原和非水溶性抗原按质量比2:1混合,即为制备粒子的抗原原料来源。1.5×10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse, the mice were sacrificed when the tumors grew to a volume of about 1000 mm 3 , and tumor tissues were excised. The tumor tissue was cut into pieces and ground, added collagenase and incubated in RPMI1640 medium for 30min, then added an appropriate amount of pure water through the cell strainer and repeated freezing and thawing 5 times, accompanied by ultrasound to destroy the lysed cells. After the cells are lysed, the lysate is centrifuged at 5000g for 5 minutes and the supernatant is taken as the water-soluble antigen soluble in pure water; add 10% sodium deoxycholate to the obtained precipitate to dissolve the precipitate. Convert the water-insoluble antigen insoluble in pure water to be soluble in 10% sodium deoxycholate aqueous solution, and mix the water-soluble antigen and the water-insoluble antigen in a mass ratio of 2:1, which is the source of antigen raw materials for preparing particles.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例中纳米粒子及作为对照的空白纳米粒采用溶剂挥发法制备,并进行了适当的修饰改进,在纳米粒子制备过程中采用低温硅化技术和添加带电物质两种修饰方法提高抗原的负载量。所采用的纳米粒子制备材料PLGA分子量为24KDa-38KDa,所采用的免疫佐剂为poly(I:C)。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载抗原和佐剂,在内部负载抗原(裂解组分)后,将100mg纳米粒子在10000g离心20分钟,然后使用7mL PBS重悬纳米粒子并与3mL含有细胞裂解物(60mg/mL)的PBS溶液混合,尔后在10000g离心20分钟,然后采用10mL硅酸盐溶液(含150mM NaCl、80mM原硅酸四甲酯和1.0mM HCl,pH3.0)重悬,并在室温固定10min,尔后在-80℃固定24h,使用超纯水离心洗涤后使用3mL含鱼精蛋白(5mg/mL)和聚赖氨酸(10mg/mL)的PBS重悬并作用10min,然后10000g离心20min洗涤,采用10mL含有裂解物(50mg/mL)的PBS溶液重悬并作用10min,然后在10000g离心20分钟并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h;在粒子使用前将其用7mL PBS重悬然后加入3mL含佐剂的癌组织裂解液组分(蛋白质浓度50mg/mL)并室温作用10min,得到内外都负载裂解物的经冷冻硅化和添加阳离子物质的修饰的纳米粒子系统。该纳米粒子平均粒径为350nm左右,纳米粒子表面电位为-3mV左右;每1mg PLGA纳米粒子约负载300μg蛋白质或多肽组分,每1mgPLGA纳米粒内外所使用的poly(I:C)免疫佐剂共约为0.02mg且内外各半。The nanoparticles in this example and the blank nanoparticles as a control were prepared by solvent evaporation method, and appropriate modification was carried out. In the preparation process of nanoparticles, two modification methods, low temperature silicidation technology and addition of charged substances were used to improve the antigen loading capacity. . The used nanoparticle preparation material PLGA has a molecular weight of 24KDa-38KDa, and the used immune adjuvant is poly(I:C). The preparation method was as described above. In the preparation process, the antigen and adjuvant were first loaded inside the nanoparticles by double emulsion method. After loading the antigen (lysed components) inside, 100 mg of nanoparticles were centrifuged at 10,000 g for 20 minutes, and then 7 mL of PBS was used. Nanoparticles were resuspended and mixed with 3 mL of PBS containing cell lysate (60 mg/mL), followed by centrifugation at 10,000 g for 20 min, followed by 10 mL of silicate solution (containing 150 mM NaCl, 80 mM tetramethyl orthosilicate, and 1.0 mM orthosilicate). HCl, pH 3.0) was resuspended and fixed at room temperature for 10 min, then fixed at -80 °C for 24 h, centrifuged with ultrapure water, washed with 3 mL of protamine (5 mg/mL) and polylysine (10 mg/mL) ) in PBS for 10min, then centrifuged at 10000g for 20min, washed with 10mL PBS solution containing lysate (50mg/mL) and acted for 10min, then centrifuged at 10000g for 20min and used 10mL supernatant containing 4% trehalose. The particles were resuspended in pure water and then freeze-dried for 48 hours; before the particles were used, they were resuspended in 7 mL of PBS, then 3 mL of adjuvanted cancer tissue lysate fraction (
对照纳米粒子将负载的癌细胞全细胞抗原替换为四种质量等量的黑色素瘤抗原多肽,其他与负载癌细胞全细胞抗原的纳米粒子相同。对照纳米粒子每1mgPLGA纳米粒所使用的poly(I:C)0.02mg,平均粒径为350nm左右,纳米粒子表面电位为-3mV左右。所负载的四种多肽新生抗原为B16-M20(Tubb3,FRRKAFLHWYTGEAMDEMEFTEAESNM),B16-M24(Dag1,TAVITPPTTTTKKARVSTPKPATPSTD),B16-M46(Actn4,NHSGLVTFQAFIDVMSRETTDTDTADQ)和TRP2:180-188(SVYDFFVWL)。The control nanoparticles replaced the loaded cancer cell whole cell antigen with four kinds of melanoma antigen polypeptides with equal mass, and others were the same as the nanoparticles loaded with cancer cell whole cell antigen. The control nanoparticles used 0.02 mg of poly(I:C) per 1 mg of PLGA nanoparticles, the average particle size was about 350 nm, and the surface potential of the nanoparticles was about -3 mV. The four peptide neoantigens loaded were B16-M20 (Tubb3, FRRKAFLHWYTGEAMDEMEFTEAESNM), B16-M24 (Dag1, TAVITPPTTTTKKARVSTPKPATPSTD), B16-M46 (Actn4, NHSGLVTFQAFIDVMSRETTDTDTADQ) and TRP2:180-188 (SVYDFFVWL).
(3)树突状细胞的制备(3) Preparation of dendritic cells
本实施例以从小鼠骨髓细胞制备树突状细胞为例来说明如何制备骨髓来源的树突状细胞(BMDC)。首先,取1只6-8周龄C57小鼠颈椎脱臼处死,手术取出后腿的胫骨和股骨放入PBS中,用剪刀和镊子将骨周围的肌肉组织剔除干净。用剪刀剪去骨头两端,再用注射器抽取PBS溶液,针头分别从骨头两端插入骨髓腔,反复冲洗骨髓到培养皿中。收集骨髓溶液,400g离心3min后加入1mL红细胞裂解液裂红。加入3mL RPMI 1640(10%FBS)培养基终止裂解,400g离心3min,弃上清。将细胞放置10mm培养皿中培养,使用RPMI 1640(10%FBS)培养基,同时加入重组小鼠GM-CSF(20ng/mL),37度,5%CO2培养7天。第3天轻轻摇晃培养瓶,补充同样体积含有GM-CSF(20ng/mL)RPMI 1640(10%FBS)培养基。第6天,对培养基进行半量换液处理。第7天,收集少量悬浮及半贴壁细胞,通过流式检测,当CD86+CD80+细胞在CD11c+细胞中的比例为15-20%之间,诱导培养的BMDC即可被用来做下一步实验。This example uses the preparation of dendritic cells from mouse bone marrow cells as an example to illustrate how to prepare bone marrow-derived dendritic cells (BMDC). First, a 6-8-week-old C57 mouse was sacrificed by cervical dislocation. The tibia and femur of the hind leg were surgically removed and placed in PBS. The muscle tissue around the bone was removed with scissors and forceps. Cut off both ends of the bone with scissors, and then use a syringe to extract the PBS solution. The needles are inserted into the bone marrow cavity from both ends of the bone, and the bone marrow is repeatedly washed into the Petri dish. The bone marrow solution was collected, centrifuged at 400 g for 3 min, and then 1 mL of erythrocyte lysate was added. 3 mL of RPMI 1640 (10% FBS) medium was added to terminate the lysis, centrifuged at 400 g for 3 min, and the supernatant was discarded. The cells were placed in a 10 mm petri dish and cultured in RPMI 1640 (10% FBS) medium with the addition of recombinant mouse GM-CSF (20 ng/mL) at 37 degrees, 5% CO 2 for 7 days. On
(4)骨髓来源巨噬细胞(BMDM)的制备(4) Preparation of bone marrow-derived macrophages (BMDM)
将C57小鼠麻醉后脱臼处死,将小鼠使用75%乙醇的消毒,然后用剪刀在小鼠背部剪开一小口,用手直接撕开皮肤至小鼠小腿关节处,去除小鼠足关节以及皮肤。用剪刀沿着小鼠大腿根部大转子将后肢拆下来,去掉肌肉组织后放置在含有75%乙醇的培养皿内浸泡5min,更换新的75%乙醇的培养皿移入超净台中。将乙醇浸泡的腿骨移入冷的PBS浸泡,洗去胫骨、股骨表面的乙醇,此过程可重复3次。将清洗好的股骨、胫骨分开,并用剪刀分别将股骨、胫骨两端剪断,使用1mL注射器吸取冷的诱导培养基将骨髓从股骨、胫骨中吹出,反复吹洗3次,直至腿骨内看不到明显的红色为止。用5mL移液枪将含有骨髓细胞的培养基反复吹打,使细胞团块分散,然后使用70μm细胞滤器将细胞过筛,转移至15mL离心管内,1500rpm/min离心5min,弃上清,加入红细胞裂解液重悬静置5min后1500rpm/min离心5min,弃上清用冷的配置好的骨髓巨噬细胞诱导培养基(含有15%L929培养基的DMEM高糖培养基)重悬,铺板。将细胞培养过夜,以去除贴壁较快的其他杂细胞如纤维细胞等等。收集未贴壁细胞按实验设计安排种入皿或细胞培养板内。巨噬细胞集落刺激因子(M-CSF)以40ng/mL浓度刺激使骨髓细胞向单核巨噬细胞分化。培养8天,光镜下观察巨噬细胞形态变化。8天后消化收集细胞,用抗小鼠F4/80抗体和抗小鼠CD11b抗体,4℃避光孵育30min后,使用流式细胞术鉴定所诱导成功的巨噬细胞的比例。The C57 mice were anesthetized and killed by dislocation. The mice were sterilized with 75% ethanol, and then a small incision was cut on the back of the mice with scissors. skin. Use scissors to remove the hindlimb along the greater trochanter at the base of the thigh of the mouse, remove the muscle tissue and place it in a petri dish containing 75% ethanol for 5 min, then replace the petri dish with a new 75% ethanol and move it into a clean bench. Transfer the ethanol-soaked leg bones into cold PBS to wash off the ethanol on the surface of the tibia and femur. This process can be repeated 3 times. Separate the cleaned femur and tibia, and cut both ends of the femur and tibia with scissors. Use a 1 mL syringe to suck out the cold induction medium to blow out the bone marrow from the femur and tibia. Repeat the blowing 3 times until the inside of the leg bone is no longer visible. to a clear red. The medium containing bone marrow cells was repeatedly pipetted with a 5mL pipette to disperse the cell clumps, then the cells were sieved using a 70μm cell strainer, transferred to a 15mL centrifuge tube, centrifuged at 1500rpm/min for 5min, the supernatant was discarded, and red blood cells were added for lysis. The solution was resuspended for 5 min and then centrifuged at 1500 rpm/min for 5 min. The supernatant was discarded and resuspended in cold prepared bone marrow macrophage induction medium (DMEM high glucose medium containing 15% L929 medium) and plated. Cells were cultured overnight to remove other fast-adhering miscellaneous cells such as fibroblasts and the like. Collect non-adherent cells and seed them into dishes or cell culture plates according to the experimental design. Macrophage colony-stimulating factor (M-CSF) was stimulated at a concentration of 40 ng/mL to differentiate myeloid cells into mononuclear macrophages. After 8 days of culture, the morphological changes of macrophages were observed under a light microscope. After 8 days, the cells were digested and collected. After incubation with anti-mouse F4/80 antibody and anti-mouse CD11b antibody for 30 min at 4°C in the dark, flow cytometry was used to identify the proportion of successfully induced macrophages.
(5)抗原提呈细胞的激活(5) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(500μg)或多肽纳米粒(500μg)与制备的BMDC(1000万个)和BMDM细胞(1000万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2),孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD40抗体(10ng/mL)。Nanoparticles (500 μg) or polypeptide nanoparticles (500 μg) loaded with cancer cell whole cell components were co-incubated with prepared BMDC (10 million) and BMDM cells (10 million) in 15 mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ), the incubation system contains granulocyte-macrophage colony stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL) , IL-12 (200U/mL) and CD40 antibody (10ng/mL).
(6)抗原提呈细胞来源的纳米疫苗的制备(6) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的BMDC和BMDM,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(22.5W)超声1分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,然后在16000g离心90分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后使用0.22μm膜过滤器挤出过滤样品。尔后将样品在混合冻干保护剂水溶液1(1%海藻糖+2%甘露醇+2%明胶)中冷冻干燥48小时即得纳米疫苗1,纳米疫苗1粒径为150纳米;或者将样品在混合冻干保护剂水溶液2(1%海藻糖+2%甘露醇+2%PVP)中冷冻干燥48小时,即得纳米疫苗2,纳米疫苗2粒径为150纳米。The incubated BMDCs and BMDMs were collected by centrifugation at 400 g for 5 min, then the cells were washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, and the cells were resuspended in PBS water at low power (22.5°C) at 4°C. W) Sonication for 1 minute. The samples were then centrifuged at 3000g for 15 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 8000g for 15 minutes, and then after 90 minutes at 16000g. The supernatant was discarded to collect the pellet and the pellet was placed in PBS After resuspension the samples were extrusion filtered using a 0.22 μm membrane filter. Then the sample was freeze-dried in mixed lyophilized protective agent aqueous solution 1 (1% trehalose + 2% mannitol + 2% gelatin) for 48 hours to obtain
(7)抗原提呈细胞制备的纳米疫苗用于癌症转移的预防(7) Nano-vaccine prepared by antigen-presenting cells for the prevention of cancer metastasis
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在小鼠癌症造模前第-35天、第-28天、第-21天、第-14天和第-7天每只小鼠皮下注射120μg纳米疫苗。在第0天给每只小鼠静脉注射接种0.5×105个B16F10细胞,第14天处死小鼠,观察记录小鼠肺部黑色素瘤癌灶数量。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. Each mouse was injected subcutaneously with 120 μg of nanovaccine on days -35, -28, -21, -14 and -7 days before cancer modeling in mice. On the 0th day, each mouse was inoculated with 0.5×10 5 B16F10 cells by intravenous injection, and the mice were sacrificed on the 14th day, and the number of melanoma tumor foci in the lungs of the mice was observed and recorded.
(8)实验结果(8) Experimental results
如图5所示,对照组小鼠的癌灶较多且癌灶较大,而纳米疫苗处理的小鼠癌症数量减少。而且,负载癌细胞全细胞抗原的纳米粒子激活的DC和巨噬细胞制备的纳米疫苗对癌症肺转移的预防效果优于负载四种抗原多肽的纳米粒子激活的DC和巨噬细胞制备的纳米疫苗。这说明负载癌细胞全细胞抗原的纳米粒子激活的抗原提呈细胞所制备的纳米疫苗能激活更广谱和多样的癌细胞特异性T细胞,因而所能识别和杀灭的癌细胞也就越多,预防癌症转移的效果也越好。而且使用混合冻干保护剂1冷冻干燥制备疫苗的效果好于使用混合冻干保护剂2冷冻干燥制备的疫苗。As shown in Figure 5, the control mice had more and larger cancer foci, while the nanovaccine-treated mice had fewer cancers. Moreover, the nanovaccine prepared by nanoparticle-activated DCs and macrophages loaded with cancer cell whole cell antigens is more effective in preventing cancer lung metastasis than the nanovaccine prepared by nanoparticle-activated DCs and macrophages loaded with four antigen polypeptides . This indicates that the nanovaccine prepared by the antigen-presenting cells activated by nanoparticles loaded with cancer cell whole cell antigens can activate a wider spectrum and variety of cancer cell-specific T cells, and thus the more cancer cells that can be recognized and killed. The more, the better the effect of preventing cancer metastasis. Moreover, the effect of freeze-drying the vaccine prepared by using the mixed lyophilized
实施例5微米粒子激活的抗原提呈细胞制备微米疫苗用于预防癌症Example 5 Micron-particle-activated antigen-presenting cells to prepare micro-vaccine for cancer prevention
本实施例中,首先使用6M盐酸胍裂解B16F10黑色素瘤癌细胞全细胞组分。全细胞抗原包含在全细胞组分中。然后,以PLGA为微米粒骨架材料,以poly(I:C)、CpGM362和CpGBW006为免疫佐剂制备负载有癌细胞全细胞抗原的微米粒子,并使用该微米粒子激活抗原提呈细胞,然后使用抗原提呈细胞制备微米疫苗用于预防癌症。In this example, the whole cell fraction of B16F10 melanoma cancer cells was firstly lysed with 6M guanidine hydrochloride. Whole cell antigens are contained in the whole cell fraction. Then, using PLGA as the microparticle skeleton material, poly(I:C), CpGM362 and CpGBW006 as immune adjuvants to prepare microparticles loaded with cancer cell whole cell antigens, and use the microparticles to activate antigen-presenting cells, and then use Antigen-presenting cells prepare micro-vaccine for cancer prevention.
(1)癌细胞的裂解(1) Lysis of cancer cells
将培养的B16F10黑色素瘤癌细胞系收集后在350g离心5分钟,然后弃去上清并用PBS洗涤两遍,然后采用6M盐酸胍重悬和裂解癌细胞,癌细胞全细胞抗原裂解并溶于6M盐酸胍后即为制备微米粒子系统的抗原原料来源。The cultured B16F10 melanoma cancer cell line was collected and centrifuged at 350g for 5 minutes, then the supernatant was discarded and washed twice with PBS, then the cancer cells were resuspended and lysed with 6M guanidine hydrochloride, and the cancer cell whole cell antigen was lysed and dissolved in 6M Guanidine hydrochloride is the source of antigen raw material for the preparation of microparticle system.
(2)微米粒子系统的制备(2) Preparation of microparticle system
本实施例中微米粒子用复乳法制备,所采用的微米粒子制备材料PLGA分子量为38KDa-54KDa,所采用的免疫佐剂为poly(I:C)、CpGM362和CpG BW006。制备方法如前所述,在制备过程中首先采用复乳法在微米粒子内部负载癌细胞全细胞抗原和佐剂,然后将100mg微米粒子在10000g离心15分钟,然后使用10mL 4%海藻糖水溶液重悬微米粒子后冷冻干燥48后备用。该微米粒子平均粒径为2.50μm左右,微米粒子表面电位为-2mV左右;每1mg PLGA微米粒子约负载100μg蛋白质或多肽组分,每1mgPLGA微米粒负载poly(I:C)、CpGM362和CpG BW006免疫佐剂各0.02mg。该微米粒子在做下述步骤(6)的癌症预防实验时定义为对照微米疫苗3。In this example, the microparticles were prepared by the double emulsion method. The molecular weight of the microparticle preparation material PLGA was 38KDa-54KDa, and the immunoadjuvants used were poly(I:C), CpGM362 and CpG BW006. The preparation method was as described above. In the preparation process, the whole-cell antigen and adjuvant of cancer cells were first loaded into the microparticles by double emulsion method, and then 100 mg of microparticles were centrifuged at 10,000 g for 15 minutes, and then resuspended with 10 mL of 4% trehalose aqueous solution. The microparticles were freeze-dried for 48 hours and used for later use. The average particle size of the microparticles is about 2.50μm, and the surface potential of the microparticles is about -2mV; each 1mg PLGA microparticle is loaded with about 100μg protein or polypeptide components, and each 1mg PLGA microparticle is loaded with poly(I:C), CpGM362 and CpG BW006 Each 0.02 mg of immune adjuvant. The microparticles were defined as the
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC和B细胞混合抗原提呈细胞。BMDC制备方法同实施例1。将小鼠处死后摘取小鼠脾脏,将脾脏剪碎后通过细胞筛网制备脾细胞单细胞悬液,然后使用磁珠分选法从脾细胞单细胞悬液中分选出CD19+B细胞。将BMDC与B细胞按照数量比1:1混合即为混合抗原提呈细胞。In this example, BMDC and B cells were mixed with antigen presenting cells. The preparation method of BMDC is the same as that in Example 1. After the mice were sacrificed, the mouse spleen was harvested, and the spleen was minced and passed through a cell mesh to prepare a single-cell suspension of spleen cells, and then CD19 + B cells were separated from the single-cell suspension of spleen cells by magnetic bead sorting. . Mixing BMDC and B cells according to the quantity ratio of 1:1 is the mixed antigen-presenting cells.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的微米粒子(500μg)与混合抗原提呈细胞(1000万个)在20mL高糖DMEM完全培养基中共孵育72小时(37℃,5%CO2)。孵育体系中含有细胞因子组分1:IL-2(500U/mL)、IL-7(200U/mL)、IL-15(200U/mL),所激活的抗原提呈细胞为被激活的混合抗原提呈细胞1;或者孵育体系中含有对照细胞因子组合2:IL-4(500U/mL)、IL-6(200U/mL)、IL-10(200U/mL),所激活的抗原提呈细胞为被激活的混合抗原提呈细胞2。Microparticles (500 μg) loaded with cancer cell whole cell fractions were co-incubated with mixed antigen-presenting cells (10 million) in 20 mL of high glucose DMEM complete medium for 72 hours (37° C., 5% CO 2 ). The incubation system contains cytokine components 1: IL-2 (500U/mL), IL-7 (200U/mL), IL-15 (200U/mL), and the activated antigen-presenting cells are activated mixed
(5)抗原提呈细胞来源的微米疫苗的制备(5) Preparation of antigen-presenting cell-derived micro-vaccine
通过在400g离心5分钟收集孵育后的混合抗原提呈细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(22.5W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,然后在16000g离心90分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后使用0.22μm膜过滤挤出即得混合抗原提呈细胞细胞膜组分。将10mL混合抗原提呈细胞细胞膜组分(4mg)与100mg步骤(2)制备的微米粒子混合,然后在4℃低功率(22.5W)超声1分钟,尔后使用匀浆机搅拌5分钟,尔后共孵育10分钟,然后在12000g离心10分钟,弃去上清液有将沉淀重悬于10mL冻干保护剂(2%海藻糖+2%甘露醇+1%精氨酸的水溶液)中冷冻干燥48小时后备用。其中,由被激活的混合抗原提呈细胞1制备的微米疫苗为微米疫苗1,粒径为2.51μm;由被激活的混合抗原提呈细胞2制备的微米疫苗为微米疫苗2,粒径为2.51μm。The incubated pooled antigen-presenting cells were collected by centrifugation at 400 g for 5 min, then washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, resuspended in PBS water at low power at 4°C (22.5W) sonicated for 2 minutes. The samples were then centrifuged at 3000g for 15 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 8000g for 15 minutes, and then after 90 minutes at 16000g. The supernatant was discarded to collect the pellet and the pellet was placed in PBS After resuspension, the mixture was filtered and extruded through a 0.22 μm membrane to obtain the cell membrane fraction of the mixed antigen-presenting cells.
(6)微米疫苗用于癌症的预防(6) Micron vaccine for cancer prevention
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在小鼠接种癌细胞前第-42天、第-35天、第-28天、第-21天、第-14天和第-7天每只小鼠分别接种100μg微米疫苗(微米疫苗1、或微米疫苗2或者步骤(2)制备的微米粒子定义为微米疫苗3)或PBS。在第0天给每只小鼠皮下注射接种1.5×105个B16F10细胞,从第3天开始每3天记录一次小鼠肿瘤体积的大小。肿瘤体积采用公式v=0.52×a×b2计算,其中v为肿瘤体积,a为肿瘤长度,b为肿瘤宽度。出于动物实验伦理,在小鼠生存期试验中当小鼠肿瘤体积超过2000mm3即视为小鼠死亡并将小鼠安乐死。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. Each mouse was vaccinated with 100 μg of the micro-vaccine (
(7)实验结果(7) Experimental results
如图6所示,PBS对照组小鼠的肿瘤都很快长大,小鼠很快死亡。而使用微米疫苗处理的小鼠肿瘤生长速度都明显变慢且生存期明显延长。而且,使用细胞因子组合1激活的混合抗原提呈细胞1制备的微米疫苗1效果明显好于使用细胞因子组分2激活的混合抗原提呈细胞2制备的微米疫苗2。而且,微米疫苗2效果也明显好于只是内部负载癌细胞全细胞组分而表面不负载任何抗原提呈细胞细胞膜组分的微米疫苗3。由此可见,表面负载抗原提呈细胞细胞膜组分能够明显提高微米疫苗的疗效;而且,在微米粒子激活抗原提呈细胞时加入特定细胞因子可以提高被激活抗原提呈细胞所制备的微米疫苗的疗效。As shown in Figure 6, the tumors of the mice in the PBS control group grew rapidly, and the mice died quickly. However, the tumor growth rate of mice treated with the micro-vaccine was significantly slower and the survival time was significantly prolonged. Moreover, the effect of the
实施例6纳米疫苗用于癌症的预防Example 6 Nano-vaccine for cancer prevention
本实施例中,首先使用8M尿素裂解B16F10黑色素瘤肿瘤组织,并溶解肿瘤组织裂解物组分。然后,以PLGA为纳米粒骨架材料,以Poly(I:C)和CpG2395为免疫佐剂制备负载有癌细胞全细胞抗原的纳米粒子,使用纳米粒子激活抗原提呈细胞后利用抗原提呈细胞制备纳米疫苗用于预防癌症。In this example, B16F10 melanoma tumor tissue was firstly lysed with 8M urea, and the tumor tissue lysate fraction was dissolved. Then, using PLGA as nanoparticle skeleton material, using Poly(I:C) and CpG2395 as immune adjuvants to prepare nanoparticles loaded with cancer cell whole cell antigens, using nanoparticles to activate antigen-presenting cells and then using antigen-presenting cells to prepare Nanovaccine for cancer prevention.
(1)肿瘤组织的收集及裂解(1) Collection and lysis of tumor tissue
在每只C57BL/6小鼠背部皮下接种1.5×105个B16F10细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,通过细胞过滤网加入适量8M尿素裂解细胞,并溶解细胞裂解物。以上即为制备纳米粒子系统的抗原原料来源。1.5×10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse, the mice were sacrificed when the tumors grew to a volume of about 1000 mm 3 , and tumor tissues were excised. The tumor tissue was cut into pieces and ground, and an appropriate amount of 8M urea was added to lyse the cells through a cell strainer, and the cell lysate was dissolved. The above is the source of antigen raw materials for the preparation of nanoparticle systems.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒子及作为对照的空白纳米粒采用溶剂挥发法制备。所采用的纳米粒子制备材料PLGA分子量为7KDa-17KDa,所采用的免疫佐剂为poly(I:C)和CpG2395,且裂解物组分和佐剂包载于纳米粒子内部。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解物组分和佐剂,然后将100mg纳米粒子在12000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h,得冻干粉后备用。该纳米粒子平均粒径为270nm左右,纳米粒子表面电位为-3mV左右;每1mg PLGA纳米粒子约负载110μg蛋白质或多肽组分,每1mg PLGA纳米粒所使用的poly(I:C)和CpG2395免疫佐剂各为0.02mg。The nanoparticles in this example and the blank nanoparticles as a control were prepared by solvent evaporation method. The adopted nanoparticle preparation material PLGA has a molecular weight of 7KDa-17KDa, the adopted immunoadjuvant is poly(I:C) and CpG2395, and the lysate components and adjuvant are encapsulated in the nanoparticle. The preparation method was as described above. In the preparation process, the lysate components and adjuvants were first loaded into the nanoparticles by double emulsion method, and then 100 mg of nanoparticles were centrifuged at 12,000 g for 20 minutes, and 10 mL of ultrapure 4% trehalose was used. After resuspension in water, freeze-dry for 48 hours, and obtain freeze-dried powder for later use. The average particle size of the nanoparticle is about 270nm, and the surface potential of the nanoparticle is about -3mV; each 1mg PLGA nanoparticle is loaded with about 110μg protein or polypeptide components, and the poly(I:C) and CpG2395 used in each 1mg PLGA nanoparticle are immune Adjuvants were each 0.02 mg.
对照纳米粒子制备材料和制备方法相同,每1mgPLGA纳米粒负载poly(I:C)和CpG2395各0.02mg,粒径为270nm左右,纳米粒子表面电位为-3mV左右,每1mg PLGA纳米粒子约负载110μg多肽组分。所负载的四种多肽新生抗原为B16-M20(Tubb3,FRRKAFLHWYTGEAMDEMEFTEAESNM),B16-M24(Dag1,TAVITPPTTTTKKARVSTPKPATPSTD),B16-M46(Actn4,NHSGLVTFQAFIDVMSRETTDTDTADQ)和TRP2:180-188(SVYDFFVWL)。The preparation materials and preparation methods of the control nanoparticles are the same, each 1mg PLGA nanoparticles is loaded with 0.02mg of poly(I:C) and CpG2395, the particle size is about 270nm, the surface potential of the nanoparticles is about -3mV, and each 1mg PLGA nanoparticles is loaded with about 110μg peptide components. The four peptide neoantigens loaded were B16-M20 (Tubb3, FRRKAFLHWYTGEAMDEMEFTEAESNM), B16-M24 (Dag1, TAVITPPTTTTKKARVSTPKPATPSTD), B16-M46 (Actn4, NHSGLVTFQAFIDVMSRETTDTDTADQ) and TRP2:180-188 (SVYDFFVWL).
(3)DC和B细胞的制备(3) Preparation of DC and B cells
处死C57BL/6后摘取小鼠淋巴结,制备小鼠淋巴结单细胞悬液,然后使用流式细胞术从淋巴结细胞单细胞悬液中分选出CD11c+DC和CD19+B细胞。After C57BL/6 was sacrificed, the lymph nodes of the mice were harvested to prepare a single-cell suspension of mouse lymph nodes, and then CD11c + DC and CD19 + B cells were sorted from the single-cell suspension of lymph node cells by flow cytometry.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(1mg)或多肽纳米粒(1mg)与DC(2000万个)和B细胞(2000万个)在20mL高糖DMEM完全培养基中共孵育72小时(37℃,5%CO2),或者将负载癌细胞全细胞组分的纳米粒子(500μg)与DC(1000万个)在20mL高糖DMEM完全培养基中共孵育72小时(37℃,5%CO2);孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD86抗体(10ng/mL)。Nanoparticles (1 mg) or polypeptide nanoparticles (1 mg) loaded with whole cell components of cancer cells were co-incubated with DC (20 million) and B cells (20 million) in 20 mL of high glucose DMEM complete medium for 72 hours (37 °C, 5% CO 2 ), or co-incubate nanoparticles (500 μg) loaded with cancer cell whole cell fractions with DCs (10 million) in 20 mL of high glucose DMEM complete medium for 72 hours (37 ° C, 5% CO 2 ) . ); the incubation system contains granulocyte-macrophage colony stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL) ) and CD86 antibody (10ng/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC和B细胞(DC细胞1000万个+B细胞1000万个)或者只收集DC细胞(2000万个),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃使用匀浆机在2000rpm搅拌破坏处理25分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,然后将所得上清液与步骤3所制备的相对应的激活抗原提呈细胞的纳米粒子(30mg)混合后在50W超声3分钟后使用0.45μm的膜过滤挤出,然后在12000g离心30分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗,纳米疫苗粒径为310nm。Incubated DC and B cells (10 million DC cells + 10 million B cells) or only DC cells (20 million) were collected by centrifugation at 400 g for 5 min, followed by 4°C phosphate buffer containing protease inhibitors The cells were washed twice with the solution (PBS), and the cells were resuspended in PBS water and disrupted with a homogenizer at 2000 rpm for 25 minutes at 4°C. Then the sample was centrifuged at 3000g for 15 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 8000g for 15 minutes, and then the obtained supernatant was compared with the corresponding activated antigen-presenting cells prepared in
(6)抗原提呈细胞来源的纳米疫苗用于癌症的预防(6) Antigen-presenting cell-derived nanovaccine for cancer prevention
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在小鼠接种癌细胞前第-42天、第-28天和第-14天给每只小鼠分别皮下注射40μg纳米疫苗或PBS。在第0天给每只小鼠皮下注射接种1.5×105个B16F10细胞,小鼠肿瘤体积和生存期监测方法同实施例5。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. Each mouse was injected subcutaneously with 40 μg of nanovaccine or PBS on days -42, -28 and -14 days before inoculation with cancer cells. On
(7)实验结果(7) Experimental results
如图7所示,对照组小鼠的肿瘤都很快长大,而经纳米疫苗处理的小鼠肿瘤生长速度都明显变慢或肿瘤消失。而且,负载癌细胞全细胞抗原的纳米粒激活的DC和B细胞制备的纳米疫苗的预防效果优于负载四种抗原多肽的纳米粒子激活的DC和B细胞制备的纳米疫苗。而且,负载癌细胞全细胞抗原的纳米粒子激活的DC和B细胞混合制备的纳米疫苗效果优于负载癌细胞全细胞抗原的纳米粒子激活的DC制备的纳米疫苗,说明多种被纳米粒子激活的抗原提呈细胞制备的纳米疫苗效果更好。这可能是因为激活的B细胞内的一部分组分包含进抗原提呈细胞制备的纳米疫苗后可以辅助增强纳米疫苗激活癌细胞特异性T细胞。As shown in Figure 7, the tumors of the control mice grew rapidly, while the tumors of the mice treated with the nanovaccine significantly slowed down or disappeared. Moreover, the preventive effect of the nanovaccine prepared from DCs and B cells activated by nanoparticles loaded with cancer cell whole cell antigens was better than that prepared by nanoparticles activated DCs and B cells loaded with four antigen polypeptides. Moreover, the nanovaccine prepared by the mixture of DCs and B cells activated by nanoparticles loaded with cancer cell whole cell antigens is more effective than the nanovaccine prepared by DCs activated by nanoparticles loaded with cancer cell whole cell antigens, indicating that a variety of nanoparticles activated The nano-vaccine prepared by antigen-presenting cells is more effective. This may be because some of the components in the activated B cells can help enhance the activation of cancer cell-specific T cells by the nanovaccine after being included in the nanovaccine prepared by antigen-presenting cells.
实施例7抗原提呈细胞制备的纳米疫苗用于治疗结肠癌Example 7 Nano-vaccine prepared by antigen-presenting cells is used to treat colon cancer
本实施例以MC38小鼠结肠癌为癌症模型来说明如何使用抗原提呈细胞制备的纳米疫苗用于治疗结肠癌。首先裂解结肠癌肿瘤组织和肺癌癌细胞以制备水溶性抗原混合物(质量比1:1)和非水溶性抗原(质量比1:1)混合物,并将水溶性抗原混合物和非水溶性抗原混合物按质量比1:1混合。然后,以PLA为纳米粒骨架材料,以CpGSL03和卡介苗(BCG)为免疫佐剂制备纳米粒子,并用该纳米粒子体外激活抗原提呈细胞后制备纳米疫苗治疗结肠癌。This example uses MC38 mouse colon cancer as a cancer model to illustrate how to use the nanovaccine prepared from antigen-presenting cells to treat colon cancer. First, colon cancer tumor tissue and lung cancer cancer cells were lysed to prepare a mixture of water-soluble antigens (mass ratio 1:1) and water-insoluble antigens (mass ratio 1:1). Mass ratio 1:1 mix. Then, nanoparticles were prepared with PLA as nanoparticle skeleton material, CpGSL03 and Bacille Calmette-Guérin (BCG) as immune adjuvants, and the nanoparticles were used to activate antigen-presenting cells in vitro to prepare a nanovaccine to treat colon cancer.
(1)肿瘤组织和癌细胞的裂解及各组分的收集(1) Lysis of tumor tissue and cancer cells and collection of components
在每只C57BL/6小鼠背部皮下接种2×106个MC38细胞在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,通过细胞过滤网加入适量纯水并反复冻融5次,并可伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以大于5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性抗原;在所得沉淀部分中加入5%十二烷基苯磺酸钠(SDS)水溶液溶解沉淀部分即可将不溶于纯水的非水溶性抗原转化为水溶液中可溶。2×10 6 MC38 cells were inoculated subcutaneously on the back of each C57BL/6 mouse. The mice were sacrificed when the tumors grew to a volume of about 1000 mm 3 , respectively, and the tumor tissue was excised. The tumor tissue was cut into pieces and ground, and an appropriate amount of pure water was added through a cell strainer and freeze-thawed for 5 times. Ultrasound was also used to destroy the lysed cells. After the cells are lysed, the lysate is centrifuged for 5 minutes at a rotating speed greater than 5000g and the supernatant is taken as the water-soluble antigen soluble in pure water; 5% sodium dodecylbenzenesulfonate ( The SDS) aqueous solution dissolves the precipitated part to convert the water-insoluble antigen that is insoluble in pure water into soluble in the aqueous solution.
将培养的LLC肺癌细胞系收集后在350g离心5分钟,然后弃去上清并用PBS洗涤两遍,然后采用超纯水重悬细胞并反复冻融5次,并可伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以3000g的转速离心6分钟并取上清液即为可溶于纯水的水溶性抗原;在所得沉淀部分中加入5%SDS水溶液溶解沉淀部分即可将不溶于纯水的非水溶性抗原转化为在SDS水溶液中可溶。The cultured LLC lung cancer cell line was collected and centrifuged at 350g for 5 minutes, then the supernatant was discarded and washed twice with PBS, then the cells were resuspended in ultrapure water and freeze-thawed 5 times, and sonicated to destroy the lysed cells . After the cells were lysed, the lysate was centrifuged at 3000g for 6 minutes, and the supernatant was taken to be the water-soluble antigen soluble in pure water; adding 5% SDS aqueous solution to the obtained precipitate to dissolve the precipitate, the insoluble antigen was Water-insoluble antigens in pure water are converted to soluble in aqueous SDS.
将来自结肠癌肿瘤组织的和肺癌癌细胞的的水溶性抗原按质量比1:1混合;溶解于5%SDS中的非水溶性抗原也按质量比1:1混合。然后将水溶性抗原混合物和非水溶性抗原混合物按照质量比1:1混合,该混合物为制备纳米粒子的原料来源。The water-soluble antigens from colon cancer tumor tissue and lung cancer cancer cells were mixed in a mass ratio of 1:1; the water-insoluble antigens dissolved in 5% SDS were also mixed in a mass ratio of 1:1. Then, the water-soluble antigen mixture and the water-insoluble antigen mixture are mixed according to a mass ratio of 1:1, and the mixture is a raw material source for preparing nanoparticles.
(2)BCG的裂解和各组分的收集(2) Cleavage of BCG and collection of components
BCG的裂解方法和各组分的收集方法同癌细胞的裂解方法和各组分的收集方法,水溶性抗原和溶解的非水溶性抗原按质量比1:1混合。The lysis method of BCG and the collection method of each component are the same as the method of lysis of cancer cells and the collection method of each component, and the water-soluble antigen and the dissolved water-insoluble antigen are mixed in a mass ratio of 1:1.
(3)纳米粒子的制备(3) Preparation of nanoparticles
本实施例中纳米粒采用溶剂挥发法制备。所采用的纳米粒子制备材料PLA分子量为20KDa,所采用的免疫佐剂为CpGSL03和BCG,且佐剂同时分布于纳米粒子内部和表面。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解物混合物和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h。使用前将20mg纳米粒重悬于0.9mL PBS中,并于0.1mL含有裂解物混合物(80mg/mL)和佐剂的样品室温混合孵育5分钟后即可使用。该纳米粒子平均粒径为280nm左右,纳米粒子表面电位为-3mV左右;每1mg PLGA纳米粒子约负载140μg蛋白质或多肽组分,每1mgPLGA纳米粒含有CpGSL03和BCG免疫佐剂各0.04mg。In this example, the nanoparticles were prepared by solvent evaporation method. The molecular weight of the nanoparticle preparation material PLA is 20KDa, and the used immune adjuvants are CpGSL03 and BCG, and the adjuvants are distributed both inside and on the surface of the nanoparticles. The preparation method was as described above. In the preparation process, the lysate mixture and adjuvant were first loaded inside the nanoparticles by double emulsion method, and then 100 mg of nanoparticles were centrifuged at 10,000 g for 20 minutes, and 10 mL of ultrapure water containing 4% trehalose was used. Freeze-dried for 48h after resuspension. Before use, 20 mg of nanoparticles were resuspended in 0.9 mL of PBS and incubated with 0.1 mL of the sample containing the lysate mixture (80 mg/mL) and adjuvant for 5 minutes at room temperature before use. The average particle size of the nanoparticles is about 280nm, and the surface potential of the nanoparticles is about -3mV; each 1 mg of PLGA nanoparticles is loaded with about 140 μg of protein or polypeptide components, and each 1 mg of PLGA nanoparticles contains 0.04 mg of CpGSL03 and 0.04 mg of BCG immune adjuvant.
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
处死C57BL/6后收集小鼠外周血,从外周血中分离外周血单核细胞(PBMC),然后使用流式细胞术从PBMC中分选出CD11c+DC和CD19+B细胞。本实施例中同时使用BMDC和BMDM作为抗原提呈细胞。BMDC制备方法同实施例2,BMDM制备方法同实施例4。Peripheral blood of mice was collected after sacrifice of C57BL/6, peripheral blood mononuclear cells (PBMC) were isolated from peripheral blood, and CD11c + DC and CD19 + B cells were sorted from PBMC using flow cytometry. In this example, both BMDC and BMDM were used as antigen-presenting cells. The preparation method of BMDC is the same as that of Example 2, and the preparation method of BMDM is the same as that of Example 4.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)与外周血来源的DC(500万个)、BMDC(500万个)、BMDM(500万个)和B细胞(500万个)在20mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO2),或者将负载癌细胞全细胞组分的纳米粒子(500μg)与外周血来源的DC(1000万个)和BMDC(1000万个)在RPMI1640完全培养基中共孵育72小时(37℃,5%CO2);孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD86抗体(10ng/mL)。Nanoparticles (1000 μg) loaded with whole cell fractions of cancer cells were mixed with peripheral blood-derived DC (5 million), BMDC (5 million), BMDM (5 million) and B cells (5 million) in 20 mL RPMI1640 Co-incubated for 48 hours in complete medium (37°C, 5% CO 2 ), or nanoparticles loaded with whole cell fractions of cancer cells (500 μg) with peripheral blood-derived DCs (10 million) and BMDCs (10 million) Incubate in RPMI1640 complete medium for 72 hours (37°C, 5% CO 2 ); the incubation system contains granulocyte-macrophage colony-stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL) , IL-7 (200U/mL), IL-12 (200U/mL) and CD86 antibody (10ng/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的外周血来源的DC、B细胞及BMDM(DC500万个+B细胞500万个+BMDM500万个)或者只收集DC和B细胞(DC750万个+B细胞750万个),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在高压均质机中(5000bar)中处理5分钟。然后将样品在2000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,将上清液与步骤3所制备的相对应的纳米粒子(50mg)常温共孵育6小时后使用0.45μm的膜过滤挤出,然后在13000g离心20分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗,纳米疫苗粒径为320纳米。Peripheral blood-derived DCs, B cells, and BMDMs after incubation were collected by centrifugation at 400 g for 5 min (DC 5 million + B cells 5 million + BMDM 5 million) or only DC and B cells (DC 7.5 million + B cells 750 10,000 cells), cells were then washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, resuspended in PBS water and treated in a high pressure homogenizer (5000 bar) for 5 minutes. Then the sample was centrifuged at 2000g for 15 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 8000g for 15 minutes, and the supernatant was co-incubated with the corresponding nanoparticles (50mg) prepared in
(6)纳米疫苗用于癌症的治疗(6) Nano vaccines for cancer treatment
选取6-8周的雌性C57BL/6为模型小鼠制备结肠癌荷瘤小鼠。在第0天给每只小鼠皮下接种2×106个MC38细胞,在第4、第7天、第10天、第15天和第20天分别给每只小鼠皮下注射50μg纳米疫苗。从第3天开始每3天记录一次小鼠肿瘤体积的大小。肿瘤体积采用公式v=0.52×a×b2计算,其中v为肿瘤体积,a为肿瘤长度,b为肿瘤宽度。出于动物实验伦理,在小鼠生存期试验中当小鼠肿瘤体积超过2000mm3即视为小鼠死亡并将小鼠安乐死。Select 6-8 week old female C57BL/6 as model mice to prepare colon cancer tumor-bearing mice. Each mouse was inoculated subcutaneously with 2×10 6 MC38 cells on
(7)实验结果(7) Experimental results
如图8所示,对照组小鼠的肿瘤都很快长大,而经纳米疫苗处理的小鼠肿瘤生长速度都明显变慢或肿瘤消失。而且,纳米粒子激活的DC、B细胞和巨噬细胞混合制备的纳米疫苗效果优于纳米粒子激活的DC和B细胞制备的纳米疫苗,说明多种被纳米粒子激活的抗原提呈细胞制备的纳米疫苗效果更好。综上所述,本发明所述抗原提呈细胞制备的纳米疫苗对结肠癌具有良好的治疗效果。As shown in Figure 8, the tumors of the control mice grew rapidly, while the tumor growth rate of the mice treated with the nanovaccine was significantly slowed down or the tumors disappeared. Moreover, the nanovaccine prepared by the mixture of nanoparticle-activated DCs, B cells and macrophages is more effective than the nanovaccine prepared by nanoparticle-activated DC and B cells, indicating that a variety of nanoparticle vaccines prepared by nanoparticle-activated antigen-presenting cells Vaccines work better. In conclusion, the nano-vaccine prepared by the antigen-presenting cells of the present invention has a good therapeutic effect on colon cancer.
实施例8抗原提呈细胞制备的纳米疫苗用于治疗黑色素瘤Example 8 Nano-vaccine prepared by antigen-presenting cells is used to treat melanoma
本实施例以黑色素瘤为癌症模型来说明如何使用负载来源于黑色素瘤和肺癌肿瘤组织的癌细胞全细胞抗原的纳米粒子激活抗原提呈细胞后,使用抗原提呈细胞制备纳米疫苗并用该纳米疫苗治疗黑色素瘤。本实施例中,首先裂解B16F10黑色素瘤肿瘤组织和LLC肺癌肿瘤组织以制备肿瘤组织的水溶性抗原混合物(质量比3:1)和非水溶性抗原混合物(3:1)。以PLGA为纳米粒骨架材料,以Poly ICLC和CpG2395为免疫佐剂制备负载上述混合物的纳米粒子,然后用该纳米粒子激活抗原提呈细胞,使用抗原提呈细胞制备纳米疫苗后用于癌症治疗。This example uses melanoma as a cancer model to illustrate how to activate antigen-presenting cells using nanoparticles loaded with cancer cell whole-cell antigens derived from melanoma and lung cancer tissue, and then use antigen-presenting cells to prepare nanovaccine and use the nanovaccine Treatment of melanoma. In this example, B16F10 melanoma tumor tissue and LLC lung cancer tumor tissue were firstly lysed to prepare a water-soluble antigen mixture (mass ratio 3:1) and a water-insoluble antigen mixture (3:1) of tumor tissue. Using PLGA as the nanoparticle skeleton material and Poly ICLC and CpG2395 as immune adjuvants, the nanoparticles loaded with the above mixture were prepared, and then the nanoparticles were used to activate antigen-presenting cells, and the antigen-presenting cells were used to prepare nano-vaccine for cancer treatment.
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
在每只C57BL/6小鼠背部皮下接种1.5×105个B16F10细胞或者2×106个LLC肺癌细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织。肿瘤的裂解和各组分收集方法同实施例1。将来自黑色素瘤肿瘤组织的和来自肺癌肿瘤组织的水溶性抗原和溶解于8M尿素中的原非水溶性抗原分别按照3:1的比例混合即得各自的混合物。然后将水溶性抗原混合物和非水溶性抗原混合物按照质量比1:1混合即为制备纳米粒子的抗原来源。1.5×10 5 B16F10 cells or 2×10 6 LLC lung cancer cells were subcutaneously inoculated on the back of each C57BL/6 mouse, and the mice were sacrificed when the tumor grew to a volume of about 1000 mm 3 , respectively, and the tumor tissue was excised. The methods of tumor lysis and fraction collection were the same as in Example 1. The water-soluble antigens from melanoma tumor tissue and from lung cancer tumor tissue and the original water-insoluble antigens dissolved in 8M urea were mixed in a ratio of 3:1 to obtain the respective mixtures. Then, the water-soluble antigen mixture and the water-insoluble antigen mixture are mixed in a mass ratio of 1:1 to prepare the antigen source of the nanoparticles.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例中纳米粒子采用复乳法制备。所采用的纳米粒子制备材料PLGA分子量为24KDa-38KDa,所采用的免疫佐剂为Poly ICLC和CpG2395。制备方法如前所述。首先将裂解物组分和佐剂负载于PLGA纳米粒子内部,然后将100mg纳米粒子在10000g离心25分钟,使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h后备用。该纳米粒子平均粒径为300nm左右,纳米粒子表面电位为-5mV左右;每1mg PLGA纳米粒子约负载80μg蛋白质和多肽组分,每1mgPLGA纳米粒负载poly ICLC和CpG2395各0.04mg。In this example, the nanoparticles were prepared by the double emulsion method. The used nanoparticle preparation material PLGA has a molecular weight of 24KDa-38KDa, and the used immune adjuvants are Poly ICLC and CpG2395. The preparation method is as described above. First, lysate components and adjuvants were loaded into PLGA nanoparticles, then 100 mg of nanoparticles were centrifuged at 10,000 g for 25 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose, and freeze-dried for 48 hours before use. The average particle size of the nanoparticles is about 300nm, and the surface potential of the nanoparticles is about -5mV; each 1 mg of PLGA nanoparticles is loaded with about 80 μg of protein and polypeptide components, and each 1 mg of PLGA nanoparticles is loaded with 0.04 mg of poly ICLC and 0.04 mg of CpG2395.
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
DC为来源于外周血的DC和BMDC的混合DC。BMDC制备方法同上。处死C57BL/6后收集小鼠外周血,从外周血中分离外周血单核细胞(PBMC),然后使用流式细胞术从PBMC中分选出CD11c+DC,将BMDC和外周血来源的DC按数量比1:1混合,即为本实施例所使用的混合DC。DCs are mixed DCs derived from peripheral blood and BMDCs. The preparation method of BMDC is the same as above. After killing C57BL/6 mice, peripheral blood was collected, peripheral blood mononuclear cells (PBMCs) were isolated from peripheral blood, and CD11c + DCs were sorted from PBMCs using flow cytometry. The quantity ratio is 1:1 mixed, that is, the mixed DC used in this example.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(800μg)与混合DC(1000万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2),孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD86抗体(10ng/mL)。Nanoparticles (800 μg) loaded with cancer cell whole cell components and mixed DCs (10 million) were co-incubated in 15 mL of high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ), and the incubation system contained granulocytes - Macrophage colony stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL) and CD86 antibody (10ng/mL) ).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC(1000万个),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声1分钟后使用匀浆机在1000rpm处理3分钟。然后将样品在3000g离心10分钟并收集上清液,将上清液在8000g离心5分钟后收集上清液,将上清液与步骤(2)所制备的纳米粒子(50mg)以及DSPE-PEG-CD32单抗(50μg)混合后在100W超声处理2分钟使用0.45μm的膜过滤,然后在15000g离心30分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗1,纳米疫苗粒径为280纳米。Incubated DCs (10 million) were collected by centrifugation at 400 g for 5 min, then cells were washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, and cells were resuspended in PBS water at 4°C low Power (20 W) was sonicated for 1 minute and then treated with a homogenizer at 1000 rpm for 3 minutes. Then the sample was centrifuged at 3000g for 10 minutes and the supernatant was collected. The supernatant was centrifuged at 8000g for 5 minutes and the supernatant was collected. The supernatant was mixed with the nanoparticles (50mg) prepared in step (2) and DSPE-PEG - CD32 monoclonal antibody (50μg) was mixed, sonicated at 100W for 2 minutes, filtered through a 0.45μm membrane, and then centrifuged at 15,000g for 30 minutes to collect and discard the supernatant to collect the precipitate, and resuspend the precipitate in PBS to obtain a
通过在400g离心5分钟收集孵育后的DC(1000万个),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声1分钟后使用匀浆机在1000rpm处理3分钟。然后将样品在3000g离心10分钟并收集上清液,将上清液在8000g离心5分钟后收集上清液,将上清液与DSPE-PEG-CD32单抗(50μg)混合后在100W超声处理2分钟,然后在18000g离心90分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗2,纳米疫苗粒径为150纳米。Incubated DCs (10 million) were collected by centrifugation at 400 g for 5 min, then cells were washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, and cells were resuspended in PBS water at 4°C low Power (20 W) was sonicated for 1 minute and then treated with a homogenizer at 1000 rpm for 3 minutes. The samples were then centrifuged at 3000g for 10 minutes and the supernatant was collected. The supernatant was centrifuged at 8000g for 5 minutes and the supernatant was collected. The supernatant was mixed with DSPE-PEG-CD32 mAb (50μg) and then sonicated at 100W. 2 minutes, and then centrifuged at 18,000 g for 90 minutes to collect and discard the supernatant to collect the precipitate. After resuspending the precipitate in PBS,
(6)纳米疫苗用于癌症的治疗(6) Nano vaccines for cancer treatment
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在第0天给每只小鼠皮下接种1.5×105个B16F10细胞,在第4、第7天、第10天、第15天和第20天分别给小鼠皮下注射100μg纳米疫苗。小鼠肿瘤体积和生存期监测同上。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. Each mouse was inoculated subcutaneously with 1.5×10 5 B16F10 cells on
(7)实验结果(7) Experimental results
如图9所示,PBS对照组小鼠的肿瘤都很快长大,与对照组相比,纳米粒子激活的DC制备的纳米疫苗1和纳米疫苗2处理的小鼠肿瘤生长速度明显变慢,而且部分小鼠肿瘤消失痊愈。而且,内部负载癌细胞全细胞组分表面负载DC细胞细胞膜组分的纳米疫苗1效果明显好于只是由DC细胞膜组分制备的纳米疫苗2综上所述,本发明所述纳米疫苗对癌症具有治疗效果,而且内部负载癌细胞全细胞组分能够使得纳米疫苗效果更佳。本实施例纳米疫苗中使用CD32单克隆抗体作为主动靶向的靶头,在实际应用中也可以使用甘露糖、CD3抗体、CD56抗体、甘露聚糖、CD205单抗、CD19单抗等任何具有靶向靶细胞能力的靶头。As shown in Figure 9, the tumors of the mice in the PBS control group grew rapidly. Compared with the control group, the tumor growth rate of the mice treated with
实施例9微米疫苗用于乳腺癌的预防Example 9 micron vaccine for breast cancer prevention
本实施例以4T1小鼠三阴性乳腺癌为癌症模型来说明如何采用8M尿素溶解癌细胞全细胞抗原并制备负载有癌细胞全细胞抗原的微米粒子,并用该微米粒子激活的抗原提呈细胞制备癌症疫苗用于预防乳腺癌。This example uses 4T1 mouse triple-negative breast cancer as a cancer model to illustrate how to use 8M urea to dissolve cancer cell whole cell antigen and prepare microparticles loaded with cancer cell whole cell antigen, and use the microparticles to activate antigen-presenting cells to prepare Cancer vaccines are used to prevent breast cancer.
(1)癌细胞的裂解(1) Lysis of cancer cells
将培养的4T1细胞在400g离心5分钟,然后用PBS洗涤两遍后重悬于超纯水中。所得癌细胞分别采用紫外线和高温加热进行灭活和变性处理,然后采用适量8M尿素裂解乳腺癌细胞并溶解裂解物即为制备粒子系统的原料来源。The cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water. The obtained cancer cells were inactivated and denatured by ultraviolet rays and high temperature heating respectively, and then the breast cancer cells were lysed with an appropriate amount of 8M urea, and the lysate was dissolved as the raw material source for preparing the particle system.
(2)微米粒子系统的制备(2) Preparation of microparticle system
本实施例中制备微米粒子采用复乳法,微米粒子骨架材料PLGA分子量为38KDa-54KDa,所采用的免疫佐剂为CpG2395和Poly(I:C)。制备时先采用复乳法制备内部负载裂解物组分和佐剂的微米粒子,然后将100mg微米粒子在9000g离心20分钟,使用10mL含4%海藻糖的超纯水重悬后干燥48h后备用。该微米粒子系统平均粒径为2.18μm左右,表面电位为-6mV左右;每1mg PLGA微米粒子约负载110μg蛋白质或多肽组分,负载CpG2395和Poly(I:C)各0.03mg。In this example, the microparticles were prepared by the double emulsion method, the molecular weight of the microparticle skeleton material PLGA was 38KDa-54KDa, and the immunoadjuvants used were CpG2395 and Poly(I:C). During preparation, the microparticles loaded with lysate components and adjuvants were prepared by the double emulsion method, and then 100 mg of microparticles were centrifuged at 9000 g for 20 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose, and dried for 48 hours before use. The average particle size of the microparticle system is about 2.18 μm, and the surface potential is about -6mV; every 1 mg of PLGA microparticles is loaded with about 110 μg of protein or polypeptide components, and each of CpG2395 and Poly(I:C) is loaded with 0.03 mg.
(3)BMDC和B细胞的制备(3) Preparation of BMDC and B cells
BMDC的制备同实施例2。B细胞的制备同实施例3。The preparation of BMDC is the same as that of Example 2. The preparation of B cells was the same as in Example 3.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的微米粒子(1000μg)与BMDC(1000万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);或者将将负载癌细胞全细胞组分的微米粒子(1000μg)与B细胞(1000万个)在15mL高糖DMEM完全培养基中共孵育72小时(37℃,5%CO2);孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD86抗体(10ng/mL)。Microparticles (1000 μg) loaded with cancer cell whole cell fractions were co-incubated with BMDC (10 million) in 15 mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); Microparticles (1000 μg) of cell fractions were incubated with B cells (10 million) in 15 mL high glucose DMEM complete medium for 72 hours (37°C, 5% CO 2 ); the incubation system contained granulocyte-macrophage colonies Stimulatory factor (GM-CSF, 2000 U/mL), IL-2 (500 U/mL), IL-7 (200 U/mL), IL-12 (200 U/mL) and CD86 antibody (10 ng/mL).
(5)抗原提呈细胞来源的微米疫苗的制备(5) Preparation of antigen-presenting cell-derived micro-vaccine
通过在400g离心5分钟收集孵育后的DC(1000万个)或者B细胞(1000万个),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(10W)超声10分钟。然后将样品在依次过孔径为10μm、5μm、2μm、1μm、0.45μm和0.22μm的膜过滤后收集滤液,将滤液与步骤2制备的微米粒子(40mg)混合后在50W超声处理1分钟后共孵育10分钟,然后在9000g离心15分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得微米疫苗,微米疫苗粒径为2.20μm。Incubated DCs (10 million) or B cells (10 million) were collected by centrifugation at 400 g for 5 min, then resuspended by washing the cells twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors Low power (10 W) sonication at 4°C for 10 minutes after PBS water. Then the sample was filtered through membranes with pore sizes of 10 μm, 5 μm, 2 μm, 1 μm, 0.45 μm and 0.22 μm in turn to collect the filtrate. The filtrate was mixed with the microparticles (40 mg) prepared in
(6)抗原提呈细胞来源的微米疫苗用于癌症的预防(6) Antigen-presenting cell-derived micro-vaccine for cancer prevention
选取6-8周的雌性BALB/c为模型小鼠制备乳腺癌荷瘤小鼠。在小鼠接种癌细胞前第-35天、第-28天、第-21天、第-14天和第-7天分别给疫苗组每只小鼠接种80μg微米疫苗;PBS对照组接种100μL的PBS。在第0天给每只小鼠皮下注射接种1×106个4T1细胞,从第3天开始每3天记录一次小鼠肿瘤体积的大小。小鼠肿瘤监测方法同上。Female BALB/c aged 6-8 weeks were selected as model mice to prepare breast cancer tumor-bearing mice. On the -35th, -28th, -21st, -14th and -7th days before the mice were inoculated with cancer cells, each mouse in the vaccine group was inoculated with 80 μg of micron vaccine; the PBS control group was inoculated with 100 μL of PBS. Each mouse was inoculated with 1×10 6 4T1 cells by subcutaneous injection on
(7)实验结果(7) Experimental results
如图10所示,PBS对照组相比,微米粒激活的抗原提呈细胞制备的微米疫苗处理的小鼠肿瘤生长速度明显变慢且生存期明显延长。而且,微米粒激活的DC制备的微米疫苗效果好于微米粒激活的B细胞制备的微米疫苗。由此可见,本发明所述的微米疫苗对乳腺癌具有预防效果。As shown in Figure 10, compared with the PBS control group, the tumor growth rate of the mice treated with the micro-vaccine prepared from the microparticle-activated antigen-presenting cells was significantly slower and the survival period was significantly prolonged. Moreover, the micro-vaccine prepared by microparticle-activated DC was more effective than the micro-vaccine prepared by microparticle-activated B cells. It can be seen that the micron vaccine of the present invention has a preventive effect on breast cancer.
实施例10纳米疫苗用于癌症转移的预防Example 10 Nano-vaccine for the prevention of cancer metastasis
(1)肿瘤组织和癌细胞的裂解(1) Lysis of tumor tissue and cancer cells
收集小鼠B16F10黑色素瘤肿瘤组织和培养的癌细胞后采用8M尿素裂解和溶解来自肿瘤组织和癌细胞的癌细胞全细胞组分,然后肿瘤组织组分和癌细胞组分按质量比1:2混溶。After collecting mouse B16F10 melanoma tumor tissue and cultured cancer cells, 8M urea was used to lyse and dissolve the cancer cell whole cell fractions from the tumor tissue and cancer cells, and then the tumor tissue fraction and cancer cell fraction were in a mass ratio of 1:2 Miscible.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例中纳米粒子采用溶剂挥发法制备,所采用的纳米粒子制备材料PLGA分子量为24KDa-38KDa,使用poly ICLC和CpG1018作为免疫佐剂。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载细胞组分和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h后备用。该纳米粒子平均粒径为250nm左右;每1mg PLGA纳米粒子约负载90μg蛋白质和多肽组分,负载polyICLC和CpG1018各0.03mg。In this example, the nanoparticles were prepared by solvent evaporation method. The molecular weight of the nanoparticle preparation material PLGA was 24KDa-38KDa, and poly ICLC and CpG1018 were used as immune adjuvants. The preparation method was as described above. In the preparation process, the cell components and adjuvants were first loaded inside the nanoparticles by double emulsion method, and then 100 mg of nanoparticles were centrifuged at 10,000 g for 20 minutes, and 10 mL of ultrapure water containing 4% trehalose was used. After resuspension, freeze-dried for 48h before use. The average particle size of the nanoparticles is about 250 nm; each 1 mg of PLGA nanoparticles is loaded with about 90 μg of protein and polypeptide components, and loaded with 0.03 mg each of polyICLC and CpG1018.
(3)DC的制备(3) Preparation of DC
本实施例使用BMDC和外周血DC的混合DC。BMDC制备方法同实施例2。外周血DC制备方法为将小鼠处死后收集小鼠外周血,然后分离小鼠PBMC,然后使用流式细胞术从小鼠PBMC中分离CD11c+的DC,将来自外周血的DC和BMDC按照数量比1:1混合。This example uses a mixed DC of BMDC and peripheral blood DC. The preparation method of BMDC is the same as that in Example 2. The preparation method of peripheral blood DC is to collect the peripheral blood of the mice after killing the mice, then separate the mouse PBMCs, and then use flow cytometry to separate the CD11c + DCs from the mouse PBMCs. 1:1 mix.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)和DC(1000万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-15(200U/mL)和CD86抗体(10ng/mL)。Nanoparticles (1000μg) and DCs (10 million) loaded with cancer cell whole cell components were co-incubated in 15mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contained granulocytes- Macrophage colony stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-15 (200U/mL) and CD86 antibody (10ng/mL) .
(4)抗原提呈细胞来源的纳米疫苗的制备(4) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的外周血来源的DC和BMDC,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤混合细胞两遍,将细胞重悬在PBS水中后在4℃和20W超声5分钟。然后将样品在3000g离心5分钟并收集上清液,将上清液在8000g离心15分钟后弃去沉淀并收集上清液后在18000g离心90分钟后使用0.22μm滤膜过滤,弃去上清液后将沉淀重悬于PBS中即得抗原提呈细胞细胞膜组分,然后将10mL该细胞膜组分(5mg)与100mg步骤(2)制备的纳米粒子混合后在室温共孵育15分钟然后通过0.45μm滤膜过滤挤出,然后在14000g离心25分钟,即得表面负载抗原提呈细胞细胞膜组分的纳米疫苗。将上述纳米疫苗使用10mL冻干保护剂1(2%海藻糖+2%甘露醇+1%精氨酸水溶液)重悬后冷冻干燥48小时所得纳米疫苗为纳米疫苗1,粒径为260nm;将上述纳米疫苗使用10mL冻干保护剂2(2%海藻糖+2%甘露醇+1%甘氨酸)重悬后冷冻干燥48小时所得纳米疫苗为纳米疫苗2,粒径为260nm。将纳米疫苗1和纳米疫苗2在室温储存360天后使用。After incubation, peripheral blood-derived DCs and BMDCs were collected by centrifugation at 400 g for 5 min, then the mixed cells were washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, and the cells were resuspended in PBS water at 4 °C and 20W sonication for 5 min. The samples were then centrifuged at 3000g for 5 minutes and the supernatant was collected. The supernatant was centrifuged at 8000g for 15 minutes and the precipitate was discarded and the supernatant was collected. After centrifugation at 18000g for 90 minutes, the supernatant was filtered with a 0.22 μm filter and the supernatant was discarded. Then, 10 mL of the cell membrane fraction (5 mg) was mixed with 100 mg of the nanoparticles prepared in step (2), incubated at room temperature for 15 minutes, and then passed through 0.45 Filter and extrude with a μm filter membrane, and then centrifuge at 14,000 g for 25 minutes to obtain a nano-vaccine with surface-loaded antigen-presenting cell membrane components. The above-mentioned nano-vaccine was resuspended with 10 mL of lyophilized protective agent 1 (2% trehalose+2% mannitol+1% arginine aqueous solution) and then freeze-dried for 48 hours. The obtained nano-vaccine was Nano-
将新冻干好的纳米疫苗1与在室温储存360天后的纳米疫苗1分别分析纳米粒子粒径,结果显示,在室温储存360天后,纳米疫苗1粒径为260nm,说明纳米疫苗1稳定性良好。此外,分别使用新制备的纳米疫苗1和在室温储存360天后的纳米疫苗1做预防癌症转移的疗效分析。The newly lyophilized
(5)纳米疫苗用于癌症转移的预防(5) Nano-vaccine for the prevention of cancer metastasis
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在小鼠接种癌细胞前第-42天、第-35天、第-28天、第-21天、第-14天和第-7天给每只小鼠分别接种100μg纳米疫苗(新制备的纳米疫苗1、或者在室温储存360天后的纳米疫苗1、或者在室温储存360天后的纳米疫苗2)或者PBS。在第0天给每只小鼠静脉注射接种0.5×105个B16F10细胞,第14天处死小鼠,观察记录小鼠肺部黑色素瘤癌灶数量。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. Each mouse was vaccinated with 100 μg of nanovaccine (freshly prepared on days -42, -35, -28, -21, -14 and -7 days before inoculation with cancer cells)
(6)实验结果(6) Experimental results
如图11所示,纳米粒激活的DC制备的纳米疫苗可以有效的预防癌症转移。而且,在室温放置360天后,纳米疫苗性质稳定,效果一致。而且,使用2%海藻糖+2%甘露醇+1%精氨酸作为冻干保护剂制备的纳米疫苗1效果好于使用2%海藻糖+2%甘露醇+1%甘氨酸作为冻干保护剂制备的纳米疫苗2;在室温储存360天后的纳米疫苗1与新制备的纳米疫苗1疗效无显著性差异,这说明,本发明所述的纳米疫苗可以保持长期稳定,冷冻干燥过程可能会破坏疫苗的结构使得疫苗不稳定以及活性降低。本发明所述特定冻干保护剂能够提高所制备纳米疫苗经历冷冻干燥以及长期保存时的疗效。As shown in Figure 11, the nanovaccine prepared from nanoparticle-activated DCs can effectively prevent cancer metastasis. Moreover, after being placed at room temperature for 360 days, the properties of the nanovaccine are stable and the effect is consistent. Moreover, the effect of
实施例11纳米疫苗用于胰腺癌的治疗Example 11 Nano-vaccine for the treatment of pancreatic cancer
本实施例中,将小鼠Pan02胰腺癌肿瘤组织和MC38结肠癌肿瘤组织裂解组分按3:1的比例负载于纳米粒子,并使用该纳米粒子激活的抗原提呈细胞制备纳米疫苗用于治疗胰腺癌。实验中,先取得小鼠胰腺癌和结肠癌肿瘤组织并将其裂解以制备水溶性抗原和溶于6M盐酸胍中的原非水溶性抗原。在制备粒子时,以PLGA为纳米粒子骨架材料,以BCG为佐剂制备纳米粒子,然后使用该纳米粒子激活抗原提呈细胞后制备纳米疫苗。In this example, mice Pan02 pancreatic cancer tumor tissue and MC38 colon cancer tumor tissue split fractions were loaded on nanoparticles at a ratio of 3:1, and the nanoparticle-activated antigen-presenting cells were used to prepare nanovaccine for treatment Pancreatic cancer. In the experiment, mouse pancreatic cancer and colon cancer tumor tissues were obtained and lysed to prepare water-soluble antigens and original water-insoluble antigens dissolved in 6M guanidine hydrochloride. When preparing the particles, PLGA is used as the nanoparticle skeleton material, and BCG is used as the adjuvant to prepare the nanoparticles, and then the nanoparticle is used to activate the antigen-presenting cells to prepare the nanovaccine.
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
在每只C57BL/6小鼠腋下皮下接种2×106个MC38结肠癌细胞或接种1×106个Pan02胰腺癌细胞,在各只小鼠所接种肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织。经肿瘤组织机械破碎后,在含5mg/mL IV型胶原酶、5mg/mL I型胶原酶、1.5mg/mL透明质酸酶以及1.5mg/mL DNA酶的RPMI1640完全培养基中37℃消化20min,收集细胞溶液并用300目灭菌尼龙网过滤去除小碎片。随后用CD45磁珠经过阴性分选得到较纯的肿瘤细胞。将所得肿瘤细胞在超纯水中反复冻融3次,并辅以超声裂解肿瘤细胞,然后在3000g离心5分钟后上清液部分即为水溶性组分,沉淀部分使用6M盐酸胍溶解后即为非水溶性组分。水溶性组分为胰腺癌肿瘤组织水溶性组分和结肠癌肿瘤组织水溶性组分3:1的混合物;非水溶性组分为胰腺癌肿瘤组织非水溶性组分和结肠癌肿瘤组织非水溶性组分3:1的混合物。水溶性组分混合物和非水溶性组分混合物按质量比1:1混合。BCG的裂解和溶解方法同肿瘤组织裂解方法,水溶性组分和非水溶性组分按质量比1:1混合。全细胞水溶性组分中含有全细胞水溶性抗原,全细胞非水溶性组分组分中含有全细胞非水溶性抗原。Each C57BL/6 mouse was inoculated subcutaneously with 2 x 10 6 MC38 colon cancer cells or 1 x 10 6 Pan02 pancreatic cancer cells in the armpit, when the inoculated tumors in each mouse grew to a volume of approximately 1000 mm, respectively Mice were sacrificed and tumor tissues were excised. After the tumor tissue was mechanically disrupted, it was digested in RPMI1640 complete medium containing 5 mg/mL type IV collagenase, 5 mg/mL type I collagenase, 1.5 mg/mL hyaluronidase and 1.5 mg/mL DNase for 20 min at 37°C , collect the cell solution and filter through 300 mesh sterile nylon mesh to remove small debris. Then, pure tumor cells were obtained by negative sorting with CD45 magnetic beads. The obtained tumor cells were repeatedly frozen and thawed in ultrapure water for 3 times, and the tumor cells were lysed by ultrasonic, and then centrifuged at 3000g for 5 minutes. It is a water-insoluble component. The water-soluble component is a 3:1 mixture of the water-soluble component of pancreatic cancer tumor tissue and the water-soluble component of colon cancer tumor tissue; the water-insoluble component is the water-insoluble component of pancreatic cancer tumor tissue and the water-insoluble component of colon cancer tumor tissue. A 3:1 mixture of sexual components. The water-soluble component mixture and the water-insoluble component mixture are mixed in a mass ratio of 1:1. The method of lysing and dissolving BCG is the same as that of tumor tissue, and the water-soluble component and the water-insoluble component are mixed in a mass ratio of 1:1. The whole-cell water-soluble fraction contains whole-cell water-soluble antigens, and the whole-cell water-insoluble fraction contains whole-cell water-insoluble antigens.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例中纳米粒子采用复乳法制备。所采用的纳米粒子制备材料PLGA分子量为7KDa-17KDa,所采用的免疫佐剂为BCG,且BCG包载于纳米粒子内。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解物组分和佐剂,然后将100mg纳米粒子在12000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h,得冻干粉后备用。在纳米粒子注射前将20mg纳米粒子溶于0.9mL PBS中,与0.1mL含裂解物(80mg/mL)的样品混合并在室温作用10min后使用。该纳米粒子平均粒径为260nm左右,纳米粒子表面电位为-4mV左右;每1mg PLGA纳米粒子约负载130μg蛋白质和多肽组分,每1mg PLGA纳米粒使用BCG免疫佐剂0.08mg。In this example, the nanoparticles were prepared by the double emulsion method. The used nanoparticle preparation material PLGA has a molecular weight of 7KDa-17KDa, the used immune adjuvant is BCG, and BCG is encapsulated in the nanoparticle. The preparation method was as described above. In the preparation process, the lysate components and adjuvants were first loaded into the nanoparticles by double emulsion method, and then 100 mg of nanoparticles were centrifuged at 12,000 g for 20 minutes, and 10 mL of ultrapure 4% trehalose was used. After resuspension in water, freeze-dry for 48 hours, and obtain freeze-dried powder for later use. 20 mg of nanoparticles were dissolved in 0.9 mL of PBS prior to nanoparticle injection, mixed with 0.1 mL of sample containing lysate (80 mg/mL) and used after 10 min at room temperature. The average particle size of the nanoparticles is about 260 nm, and the surface potential of the nanoparticles is about -4mV; each 1 mg of PLGA nanoparticles is loaded with about 130 μg of protein and polypeptide components, and 0.08 mg of BCG immune adjuvant is used per 1 mg of PLGA nanoparticles.
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC或者BMDM作为抗原提呈细胞。BMDC制备方法同实施例2,BMDM制备方法同实施例4。In this example, BMDC or BMDM were used as antigen-presenting cells. The preparation method of BMDC is the same as that of Example 2, and the preparation method of BMDM is the same as that of Example 4.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(1000万个)或者与BMDM(1000万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有GM-CSF(2000U/mL)、M-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)。Nanoparticles (1000 μg) loaded with cancer cell whole cell fractions were co-incubated with BMDC (10 million) or with BMDM (10 million) in 15 mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ; The incubation system contains GM-CSF (2000U/mL), M-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500 U/mL) and CD80 antibody (10 ng/mL).
(4)抗原提呈细胞来源的纳米疫苗的制备(4) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的BMDC或者BMDM,然后使用含有0.0759M蔗糖和0.225M甘露醇的30mM pH 7.0Tris-HCl缓冲液中1200rpm 3min离心清洗三次,然后在磷酸酶抑制剂和蛋白酶抑制剂的存在下使用匀浆机在2000rpm作用10分钟,然后在20W超声1分钟机械破坏抗原提呈细胞。经过离心后所获细胞膜用10mM pH 7.5的Tris-HCl和1mM EDTA的溶液清洗。然后将样品依次过孔径为30μm、10μm、5μm、2μm、0.45μm的膜过滤后,将滤液在12000g离心25分钟后弃去上清液收集沉淀,将沉淀在含有的4%甘露醇的生理盐水中重悬后冷冻干燥,即得纳米疫苗,纳米疫苗粒径为260纳米。Incubated BMDCs or BMDMs were collected by centrifugation at 400 g for 5 min, then washed three times by centrifugation at 1200 rpm for 3 min in 30 mM pH 7.0 Tris-HCl buffer containing 0.0759 M sucrose and 0.225 M mannitol, and then treated with phosphatase inhibitor and protease inhibitor Antigen presenting cells were mechanically disrupted using a homogenizer at 2000 rpm for 10 minutes in the presence of the agent, followed by sonication at 20W for 1 minute. Cell membranes obtained after centrifugation were washed with a solution of 10 mM Tris-HCl pH 7.5 and 1 mM EDTA. Then, the samples were filtered through membranes with pore sizes of 30 μm, 10 μm, 5 μm, 2 μm, and 0.45 μm in turn. The filtrate was centrifuged at 12,000 g for 25 minutes, and the supernatant was discarded to collect the precipitate. The precipitate was placed in physiological saline containing 4% mannitol. The nano-vaccine is obtained after being resuspended and then freeze-dried, and the particle size of the nano-vaccine is 260 nm.
(5)纳米疫苗用于癌症的治疗(5) Nano-vaccine for cancer treatment
选取6-8周的雌性C57BL/6为模型小鼠制备胰腺癌荷瘤小鼠。在第0天给每只小鼠皮下接种1×106个Pan02细胞,在第4、第7天、第10天、第15天、第20天和第25天分别给小鼠皮下注射400μg的纳米疫苗或PBS。从第3天开始每3天记录一次小鼠肿瘤体积的大小。肿瘤体积计算方法和小鼠生存期监测方法同上。The 6-8 week old female C57BL/6 was selected as model mice to prepare pancreatic cancer tumor-bearing mice. Each mouse was inoculated subcutaneously with 1 x 106 Pan02 cells on
(6)实验结果(6) Experimental results
如图12所示,与PBS对照组相比,纳米粒激活的DC或巨噬细胞制备的纳米疫苗都可有效治疗胰腺癌,而且,使用纳米粒激活的DC制备的纳米疫苗效果更好。As shown in Figure 12, compared with the PBS control group, the nanovaccine prepared by nanoparticle-activated DCs or macrophages can effectively treat pancreatic cancer, and the nanovaccine prepared by using nanoparticle-activated DCs is more effective.
实施例12纳米疫苗用于癌症的预防Example 12 Nano-vaccine for cancer prevention
本实施例以甘露糖为靶头说明如何使用主动靶向纳米粒激活的抗原提呈细胞制备纳米疫苗并用于预防癌症。在实际应用时具体剂型,佐剂,给药时间、给药次数、给药方案可根据情况调整。纳米粒子系统可通过DC表面的甘露糖受体摄取进入树突状细胞,并进而激活DC后制备纳米疫苗用于预防癌症。This example uses mannose as the target to illustrate how to use antigen-presenting cells activated by active targeting nanoparticles to prepare nano-vaccine for cancer prevention. In actual application, the specific dosage form, adjuvant, administration time, administration frequency, and administration schedule can be adjusted according to the situation. The nanoparticle system can be taken up into dendritic cells through mannose receptors on the surface of DCs, and then activated DCs to prepare nanovaccine for cancer prevention.
(1)癌细胞的裂解(1) Lysis of cancer cells
收集培养的B16F10癌细胞后采用8M尿素裂解和溶解来源于癌细胞的癌细胞全细胞组分。After collecting the cultured B16F10 cancer cells, 8M urea was used to lyse and lyse the cancer cell whole cell fractions derived from the cancer cells.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒子系统使用复乳法制备。所采用的纳米粒子制备材料为PLGA和甘露糖修饰的PLGA,二者分子量都为7KDa-17KDa。制备带有靶头的纳米粒子时二者一起使用时质量比为4:1。所采用的免疫佐剂为Poly(I:C)和CpG SL03。制备方法如前所述,采用复乳法将裂解物组分和佐剂共负载于纳米粒子内部,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h后备用。带有靶头纳米粒子的平均粒径均为270nm左右,每1mg PLGA纳米粒子约负载80μg蛋白质和多肽组分,含Poly(I:C)和CpGSL03各0.04mg。不负载佐剂但带有甘露糖靶头的对照纳米粒粒径也为270nm左右,制备时采用等量细胞组分但是不含任何免疫佐剂,每1mg PLGA纳米粒子约负载80μg蛋白质和多肽组分。带有甘露糖靶头的空白纳米粒粒径为250nm左右,制备时采用等量佐剂,但不负载任何细胞裂解组分。In this example, the nanoparticle system was prepared by the double emulsion method. The used nanoparticle preparation materials are PLGA and mannose-modified PLGA, both of which have a molecular weight of 7KDa-17KDa. The mass ratio of the two is 4:1 when used together to prepare nanoparticles with target heads. The immunoadjuvants used were Poly(I:C) and CpG SL03. The preparation method was as described previously. The lysate components and adjuvants were co-loaded inside the nanoparticles by the double emulsion method, and then 100 mg of the nanoparticles were centrifuged at 10,000 g for 20 minutes and resuspended in 10 mL of ultrapure water containing 4% trehalose. Freeze-dried for 48h for later use. The average particle size of the nanoparticles with the target head is about 270 nm, and each 1 mg of PLGA nanoparticles is loaded with about 80 μg of protein and polypeptide components, including 0.04 mg each of Poly(I:C) and CpGSL03. The particle size of the control nanoparticles without adjuvant but with mannose target is also about 270nm. The same amount of cell components are used in preparation but without any immune adjuvant. Each 1mg PLGA nanoparticles is loaded with about 80μg protein and peptide group. point. The particle size of blank nanoparticles with mannose targets is about 250 nm, and the same amount of adjuvant is used for preparation, but no cell lysis components are loaded.
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC和DC2.4作为抗原提呈细胞。BMDC制备方法同实施例2。This example used BMDC and DC2.4 as antigen presenting cells. The preparation method of BMDC is the same as that in Example 2.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)或者空白纳米粒子纳米粒子(1000μg)+游离裂解液与BMDC(500万个)及DC2.4细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有GM-CSF(2000U/mL)、M-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)。The nanoparticles loaded with whole cell components of cancer cells (1000μg) or blank nanoparticles nanoparticles (1000μg) + free lysate were mixed with BMDC (5 million) and DC2.4 cells (5 million) in 15mL of high glucose DMEM. The medium was co-incubated for 48 hours (37°C, 5% CO 2 ); the incubation system contained GM-CSF (2000U/mL), M-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200 U/mL), IL-12 (200 U/mL), IFN-γ (500 U/mL) and CD80 antibody (10 ng/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在5000g离心10分钟后收集上清液,将上清液通过0.45μm的膜过滤后使用超滤膜(截留分子量50KDa)超滤离心过滤和浓缩,将过滤和浓缩后的样品在17000g离心120分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗,纳米疫苗粒径为120纳米。Incubated DCs were collected by centrifugation at 400 g for 5 min, cells were washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, cells were resuspended in PBS water and sonicated at 4°C at low power (20W) 2 minutes. Then the sample was centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 5000g for 10 minutes and the supernatant was collected. The supernatant was filtered through a 0.45 μm membrane and then used an ultrafiltration membrane (molecular weight cut-off 50KDa) to supernatant. Filtration, centrifugal filtration and concentration, the filtered and concentrated samples were centrifuged at 17,000g for 120 minutes, collected, discarded the supernatant, collected the precipitate, and resuspended the precipitate in PBS to obtain a nano-vaccine with a particle size of 120 nm.
(6)纳米疫苗用于癌症的预防(6) Nano vaccines for cancer prevention
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠,在小鼠接种癌细胞前第35天、第28天、第21天、第14天和第7天分别给每只小鼠接种400μg纳米疫苗或者PBS,在第0天给每只受体小鼠背部右下方皮下接种1.5×105个B16F10细胞。监测小鼠肿瘤生长速度和小鼠生存期。肿瘤生长和生存期监测方法同上。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice, which were given on the 35th, 28th, 21st, 14th and 7th days before the mice were inoculated with cancer cells, respectively. Each mouse was inoculated with 400 μg of nanovaccine or PBS, and 1.5×10 5 B16F10 cells were subcutaneously inoculated into the lower right back of each recipient mouse on
(5)实验结果(5) Experimental results
如图13所示,与PBS对照组和主动靶向的空白纳米粒+游离裂解物激活的DC制备的纳米疫苗对照组相比,负载全细胞组分的纳米粒子激活的DC制备的纳米疫苗处理的小鼠其肿瘤生长速度明显变慢。不管是否带有佐剂,纳米粒子激活的DC制备的纳米疫苗都可以有效预防癌症,但是带有佐剂效果更佳。而且这说明本发明所述的纳米疫苗可以有效预防癌症。As shown in Figure 13, compared with the PBS control group and the nanovaccine control group prepared from actively targeted blank nanoparticles + free lysate-activated DCs, the nanovaccine treatment of nanoparticle-activated DCs loaded with whole cell components of mice had significantly slower tumor growth. With or without adjuvant, nanovaccine prepared from nanoparticle-activated DCs can effectively prevent cancer, but with adjuvant, the effect is better. And this shows that the nano-vaccine of the present invention can effectively prevent cancer.
实施例13纳米疫苗用于预防肝癌Example 13 Nano-vaccine for the prevention of liver cancer
本实施例中,首先裂解Hepa1-6肝癌细胞,以PLGA为纳米粒子骨架材料,以Poly(I:C)和BCG为免疫佐剂制备负载来源于肝癌细胞的癌细胞全细胞组分的纳米粒子,然后以该粒子激活的DC和B细胞制备纳米疫苗用于预防肝癌。In this example, Hepa1-6 liver cancer cells were firstly lysed, PLGA was used as nanoparticle skeleton material, and Poly(I:C) and BCG were used as immune adjuvants to prepare nanoparticles loaded with whole cell components of cancer cells derived from liver cancer cells , and then prepare nano-vaccine with DC and B cells activated by the particle for the prevention of liver cancer.
(1)癌细胞的裂解及各组分的收集(1) Lysis of cancer cells and collection of components
收集培养的Hepa 1-6肝癌细胞后使用PBS洗涤两遍,使用加热和紫外照射处理肝癌细胞,尔后采用8M尿素水溶液(含200MM氯化钠)裂解和溶解来源于癌细胞的癌细胞全细胞组分。使用8M尿素水溶液(含200MM氯化钠)裂解BCG后溶解裂解组分作为佐剂使用。The cultured Hepa 1-6 hepatoma cells were collected and washed twice with PBS. The hepatoma cells were treated with heating and ultraviolet irradiation, and then 8M urea aqueous solution (containing 200mM sodium chloride) was used to lyse and lyse the whole cell group of cancer cells derived from cancer cells. point. Use 8M urea aqueous solution (containing 200mM sodium chloride) to dissolve BCG and dissolve the cleavage components as an adjuvant.
(3)纳米粒子系统的制备(3) Preparation of nanoparticle system
本实施例中纳米粒子系统采用溶剂挥发法制备,所采用的纳米粒子制备材料PLGA分子量为24KDa-38KDa,所采用的免疫佐剂为BCG和Poly(I:C),佐剂包载于纳米粒子内。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载癌细胞全细胞组分和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h后备用。该纳米粒子平均粒径为285nm左右;每1mg PLGA纳米粒子约负载100μg蛋白质和多肽组分,BCG和Poly(I:C)各0.04mg。对照纳米粒子平均粒径为285nm左右,每1mg PLGA纳米粒子约负载100μg蛋白质和多肽组分,不负载任何佐剂。In this example, the nanoparticle system is prepared by solvent evaporation method, the molecular weight of the nanoparticle preparation material PLGA is 24KDa-38KDa, the adopted immune adjuvant is BCG and Poly(I:C), and the adjuvant is encapsulated in the nanoparticle Inside. The preparation method was as described above. In the preparation process, the whole cell components of cancer cells and adjuvants were first loaded into the nanoparticles by the double emulsion method, and then 100 mg of the nanoparticles were centrifuged at 10,000 g for 20 minutes, and 10 mL of 4% trehalose was used. Resuspend in ultrapure water and freeze-dry for 48 h before use. The average particle size of the nanoparticles is about 285 nm; each 1 mg of PLGA nanoparticles is loaded with about 100 μg of protein and polypeptide components, and each of BCG and Poly(I:C) is 0.04 mg. The average particle size of the control nanoparticles was about 285 nm, and each 1 mg of PLGA nanoparticles loaded about 100 μg of protein and polypeptide components without any adjuvant.
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC和B作为抗原提呈细胞。BMDC制备方法同实施例2。B细胞来自小鼠外周血PBMC,制备方法同上。This example used BMDC and B as antigen presenting cells. The preparation method of BMDC is the same as that in Example 2. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)和CD40抗体(20mg/mL)。Nanoparticles (1000 μg) loaded with cancer cell whole cell fractions were co-incubated with BMDC (5 million) and B cells (5 million) in 15 mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ; The incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500U/mL) and CD80 antibody (10 ng/mL) and CD40 antibody (20 mg/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC和B细胞(500万个DC+500万个B细胞),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在5000g离心10分钟后收集上清液,将上清液通过0.45μm的膜过滤后使用超滤膜(截留分子量50KDa)超滤离心过滤和浓缩,将过滤和浓缩后的样品与步骤2制备的纳米粒子(10mg)混合后使用高压均质机(10000bar)处理3分钟后通过0.45μm的滤膜过滤,然后在13000g离心30分钟后弃去上清液收集沉淀,将沉淀在混合冻干保护剂水溶液(2%海藻糖+2%甘露醇+1%白蛋白)中重悬,尔后冷冻干燥48小时后即得纳米疫苗,其粒径为300纳米。After incubation, DCs and B cells (5 million DC + 5 million B cells) were collected by centrifugation at 400 g for 5 min, and then washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors. After resuspension in PBS water, low power (20W) sonication was performed at 4°C for 2 minutes. Then the sample was centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 5000g for 10 minutes and the supernatant was collected. The supernatant was filtered through a 0.45 μm membrane and then used an ultrafiltration membrane (molecular weight cut-off 50KDa) to supernatant. Filtration and centrifugal filtration and concentration, the filtered and concentrated sample was mixed with the nanoparticles (10 mg) prepared in
(4)肝癌的预防(4) Prevention of liver cancer
选取6-8周的雌性C57BL/6为模型小鼠制备肝癌荷瘤小鼠。在小鼠接种癌细胞前第-42天、第-35天、第-28天、第-21天和第-7天分别注射80μg纳米疫苗或者PBS。同时在第0天给每只小鼠皮下注射接种1.0×106个Hepa1-6肝癌细胞,肿瘤生长和小鼠生存期记录方式同上。The 6-8 week old female C57BL/6 was selected as the model mice to prepare liver cancer tumor-bearing mice. The mice were injected with 80 μg of nanovaccine or PBS on days -42, -35, -28, -21 and -7 days before inoculation of cancer cells. At the same time, 1.0×10 6 Hepa1-6 hepatoma cells were subcutaneously injected into each mouse on
(5)实验结果(5) Experimental results
如图14所示,与PBS对照组相比,纳米粒辅激活的抗原提呈细胞制备的纳米疫苗处理的小鼠肿瘤生长速度明显变慢。而且,不管是否带有佐剂,纳米粒子激活的抗原提呈细胞制备的纳米疫苗都可以有效预防癌症,但是带有佐剂的效果更好。这说明本发明所述的纳米疫苗可以有效预防癌症。As shown in Figure 14, compared with the PBS control group, the tumor growth rate of the mice treated with the nanovaccine prepared from the nanoparticle co-activated antigen-presenting cells was significantly slower. Moreover, nanovaccine prepared by nanoparticle-activated antigen-presenting cells can effectively prevent cancer with or without adjuvant, but the effect with adjuvant is better. This shows that the nanovaccine of the present invention can effectively prevent cancer.
实施例14抗原提呈细胞制备的纳米疫苗用于预防癌症Example 14 Nano-vaccine prepared by antigen-presenting cells for cancer prevention
(1)肿瘤组织和癌细胞的裂解(1) Lysis of tumor tissue and cancer cells
收集小鼠B16F10黑色素瘤肿瘤组织和培养的癌细胞后采用10%脱氧胆酸钠水溶液(含8M精氨酸)裂解和溶解来源于肿瘤组织和癌细胞的癌细胞全细胞组分,然后肿瘤组织组分和癌细胞组分按质量比1:1混溶。After collecting mouse B16F10 melanoma tumor tissue and cultured cancer cells, 10% sodium deoxycholate aqueous solution (containing 8M arginine) was used to lyse and dissolve the whole cell components of cancer cells derived from tumor tissue and cancer cells, and then tumor tissue The components and the cancer cell components were miscible at a mass ratio of 1:1.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例中纳米粒子采用溶剂挥发法制备,所采用的纳米粒子制备材料PLGA分子量为7KDa-17KDa,所采用免疫佐剂为CpG2006、CpG1018和Poly(I:C),增加溶酶体逃逸物质为精氨酸,免疫佐剂和精氨酸均负载于纳米粒子内部。制备方法如前所述,采用复乳法在纳米粒子内部负载抗原、精氨酸和佐剂,然后将100mg PLGA纳米粒子在13000g离心20min后弃去上清液并收集沉淀,将沉淀重悬于4%海藻糖中,冷冻干燥48小时后备用。该纳米粒子平均粒径为240nm左右;每1mg PLGA纳米粒子约负载80μg蛋白质或多肽组分,负载CpG2006、CpG1018和Poly(I:C)各0.03mg,负载精氨酸0.02mg。In this example, the nanoparticles are prepared by solvent evaporation method, the molecular weight of the nanoparticle preparation material PLGA is 7KDa-17KDa, the immune adjuvants used are CpG2006, CpG1018 and Poly(I:C), and the lysosome escape substances are Arginine, immune adjuvant and arginine were all loaded inside the nanoparticles. The preparation method was as described above. Antigen, arginine and adjuvant were loaded inside the nanoparticles by double emulsion method, and then 100 mg of PLGA nanoparticles were centrifuged at 13000 g for 20 min, the supernatant was discarded, and the precipitate was collected, and the precipitate was resuspended in 4 . % trehalose, freeze-dried for 48 hours before use. The average particle size of the nanoparticles is about 240 nm; each 1 mg of PLGA nanoparticles is loaded with about 80 μg of protein or polypeptide components, 0.03 mg of CpG2006, CpG1018 and Poly(I:C), and 0.02 mg of arginine.
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC和B作为抗原提呈细胞。BMDC制备方法同实施例2。B细胞来自小鼠外周血PBMC,制备方法同上。This example used BMDC and B as antigen presenting cells. The preparation method of BMDC is the same as that in Example 2. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)和CD40抗体(20mg/mL)。Nanoparticles (1000 μg) loaded with cancer cell whole cell fractions were co-incubated with BMDC (5 million) and B cells (5 million) in 15 mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ; The incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500U/mL) and CD80 antibody (10 ng/mL) and CD40 antibody (20 mg/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(10W)超声10分钟。随后将样品依次通过2μm、1μm、0.45μm和0.22μm的膜过滤后收集滤液,然后将滤液在15000g离心30分钟后收集弃去上清液收集沉淀在PBS中重悬即为抗原提呈细胞细胞膜组分。然后10mL抗原提呈细胞细胞膜组分(5mg)与100mg步骤(2)制备的纳米粒子混合后在室温在低功率(20W)超声处理2分钟后共孵育10分钟,然后通过0.45μm的膜过滤挤出,然后在15000g离心20分钟后弃去上清液后将沉淀在4%海藻糖水溶液中重悬,尔后冷冻干燥48小时后即得纳米疫苗2,纳米疫苗2粒径为250纳米;或者将沉淀在混合冻干保护剂水溶液(2%蔗糖+2%甘露醇+1%赖氨酸)中重悬,尔后冷冻干燥48小时后即得纳米疫苗1,纳米疫苗1粒径为250纳米。将冷冻干燥后的纳米疫苗1和纳米疫苗2在室温放置180天后使用,在使用时将1mg纳米疫苗重悬于1mL的PBS中。Incubated DCs and B cells were collected by centrifugation at 400 g for 5 min, then washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, resuspended in PBS water at 4°C low power ( 10W) sonicated for 10 minutes. Then, the samples were filtered through 2 μm, 1 μm, 0.45 μm and 0.22 μm membranes in turn, and the filtrate was collected, and then the filtrate was collected after centrifugation at 15,000 g for 30 minutes. components. Then 10 mL of antigen-presenting cell membrane fraction (5 mg) was mixed with 100 mg of the nanoparticles prepared in step (2) and incubated for 10 minutes at room temperature after sonication at low power (20 W) for 2 minutes, and then filtered and squeezed through a 0.45 μm membrane. After centrifugation at 15000g for 20 minutes, the supernatant was discarded, and the precipitate was resuspended in a 4% trehalose aqueous solution, and then freeze-dried for 48 hours to obtain nano-
(4)纳米疫苗癌症的预防(4) Nano-vaccine cancer prevention
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠,在小鼠接种癌细胞前第-35天、第-28天、第-21天、第-14天和第-7天每只小鼠分别接种已经在室温放置超过180天的100μg纳米疫苗1或者100μg纳米疫苗2或者PBS。在第0天,给每只受体小鼠背部右下方皮下接种1.5×105个B16F10细胞。小鼠肿瘤生长和生存期监测方法同上。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice, on the -35th, -28th, -21st, -14th and -7 days Each mouse was vaccinated with 100 μg of
(5)实验结果(5) Experimental results
如图15所示,与PBS对照组相比,纳米粒激活的抗原提呈细胞制备的纳米疫苗1和纳米疫苗2都可以非常显著的延长小鼠生存期有效预防癌症。而且,使用混合冻干保护剂(2%蔗糖+2%甘露醇+1%赖氨酸)冷冻干燥制备得到的纳米疫苗1效果好于使用4%海藻糖冻干保护剂冷冻干燥后制备得到的纳米疫苗2。As shown in Figure 15, compared with the PBS control group, both
实施例15纳米疫苗用于黑色素瘤的治疗Example 15 Nano-vaccine for the treatment of melanoma
本实施例以小鼠黑色素瘤为癌症模型来说明如何使用纳米疫苗治疗黑色素瘤。This example uses mouse melanoma as a cancer model to illustrate how to use nanovaccine to treat melanoma.
(1)肿瘤组织和癌细胞的裂解及各组分的收集(1) Lysis of tumor tissue and cancer cells and collection of components
收集肿瘤组织时先在每只C57BL/6小鼠背部皮下接种1.5×105个B16F10细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织,将肿瘤组织切块后研磨,通过细胞过滤网后制备单细胞悬液,加入超纯水后反复冻融并伴有超声裂解上述细胞,然后加入核酸酶作用5分钟,再在95℃作用10分钟灭活核酸酶。尔后在8000g离心3分钟,上清液部分即为水溶性抗原;沉淀部分使用10%脱氧胆酸钠水溶液溶解非水溶性抗原。将水溶性抗原和脱氧胆酸钠溶解后的非水溶性抗原按质量比1:1混溶即为制备纳米粒子系统的全细胞抗原原料来源。When collecting tumor tissue, firstly inoculate 1.5×10 5 B16F10 cells subcutaneously on the back of each C57BL/ 6 mouse. When the tumor grows to a volume of about 1000 mm, the mice are sacrificed and the tumor tissue is excised. The tumor tissue is cut into pieces. Grind, pass through a cell strainer to prepare a single cell suspension, add ultrapure water, freeze and thaw repeatedly and lyse the above cells with ultrasonication, then add nuclease for 5 minutes, and then inactivate nuclease at 95°C for 10 minutes. Then, centrifuge at 8000g for 3 minutes, and the supernatant part is water-soluble antigen; the precipitate part is dissolved in water-insoluble antigen with 10% sodium deoxycholate aqueous solution. The water-soluble antigen and the water-insoluble antigen after dissolving sodium deoxycholate are mixed in a mass ratio of 1:1, which is the whole cell antigen raw material source for preparing the nanoparticle system.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒采用复乳法制备,具有靶向树突状细胞的能力。所采用的纳米粒子制备材料为PLGA和甘露聚糖修饰的PLGA,二者分子量都为24KDa-38KDa,使用时未修饰PLGA和甘露聚糖修饰PLGA的质量比为9:1。所采用的免疫佐剂为poly(I:C)、CpG1018和CpG2216,增加溶酶体免疫逃逸的物质为KALA多肽(WEAKLAKALAKALAKHLAKALAKALKACEA),且佐剂、KALA多肽包载于纳米粒子内。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解液组分、佐剂、KALA多肽,在内部负载上述组分后,将100mg纳米粒子在12000g离心25分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h。该纳米粒子平均粒径为250nm左右,表面电位为-5mV左右;每1mg PLGA纳米粒子约负载100μg蛋白质或多肽组分,每1mg PLGA纳米粒所负载的poly(I:C)、CpG1018和CpG2216免疫佐剂各0.02mg,负载KALA多肽0.05mg。纳米粒2的制备材料和方法相同,其粒径为250nm左右,表面电位为-5mV左右,不负载KALA多肽,负载等量佐剂和细胞裂解组分。纳米粒子3的制备材料和制备方法相同,为250nm左右,表面电位为-5mV左右;每1mg PLGA纳米粒子约负载100μg蛋白质和多肽组分,每1mg PLGA纳米粒所负载的poly(I:C)0.02mg,负载CpG1018为0.04mg,负载KALA多肽0.05mg。In this example, the nanoparticles are prepared by the double emulsion method, and have the ability to target dendritic cells. The used nanoparticle preparation materials are PLGA and mannan-modified PLGA, both of which have molecular weights of 24KDa-38KDa, and the mass ratio of unmodified PLGA and mannan-modified PLGA is 9:1. The adopted immune adjuvants are poly(I:C), CpG1018 and CpG2216, and the substances that increase lysosomal immune escape are KALA polypeptide (WEAKLAKALAKALAKHLAKALAKALKACEA), and the adjuvant and KALA polypeptide are encapsulated in nanoparticles. The preparation method was as described above. In the preparation process, the lysate components, adjuvant, and KALA polypeptide were first loaded into the nanoparticles by the double emulsion method. And use 10mL of ultrapure water containing 4% trehalose to resuspend and freeze-dry for 48h. The average particle size of the nanoparticles is about 250nm, and the surface potential is about -5mV; each 1mg PLGA nanoparticles is loaded with about 100μg protein or polypeptide components, and the poly(I:C), CpG1018 and CpG2216 loaded per 1mg PLGA nanoparticles are immune Each adjuvant is 0.02mg, and the load of KALA polypeptide is 0.05mg. The preparation material and method of
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC和B作为抗原提呈细胞。BMDC制备方法同实施例2。B细胞来自小鼠外周血PBMC,制备方法同上。This example used BMDC and B as antigen presenting cells. The preparation method of BMDC is the same as that in Example 2. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)和CD40抗体(20mg/mL)。Nanoparticles (1000 μg) loaded with cancer cell whole cell fractions were co-incubated with BMDC (5 million) and B cells (5 million) in 15 mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ; The incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500U/mL) and CD80 antibody (10 ng/mL) and CD40 antibody (20 mg/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,将上清液通过0.22μm的膜过滤挤出,然后在16000g离心60分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗,纳米疫苗粒径为150纳米。Incubated DCs and B cells were collected by centrifugation at 400 g for 5 min, then washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, resuspended in PBS water at 4°C low power ( 20W) sonicated for 2 minutes. The samples were then centrifuged at 3000g for 15 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 8000g for 15 minutes, the supernatant was extruded by filtration through a 0.22 μm membrane, and then collected after centrifugation at 16000g for 60 minutes The supernatant was discarded to collect the precipitate, and the precipitate was resuspended in PBS to obtain a nano-vaccine. The particle size of the nano-vaccine was 150 nm.
(4)纳米疫苗用于治疗癌症(4) Nano-vaccine for the treatment of cancer
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在第0天给每只小鼠背部右下方皮下接种1.5×105个B16F10细胞。在接种黑色素瘤后第6天、第10天、第15天和第20天分别皮下40μg纳米疫苗或者PBS。在实验中,小鼠肿瘤体积和生存期监测方法同上。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. 1.5 x 105 B16F10 cells were inoculated subcutaneously in the lower right back of each mouse on
(5)实验结果(5) Experimental results
如图16所示,PBS对照组的肿瘤很快都长大。与对照组相比,纳米粒子激活的抗原提呈细胞制备的纳米疫苗处理的小鼠肿瘤生长速度明显变慢生存期明显延长。而且,加入增加溶酶体逃逸物质的纳米粒子激活的抗原提呈细胞制备的纳米疫苗好于未加入溶酶体逃逸的纳米粒子激活的抗原提呈细胞制备的纳米疫苗。而且,使用两种CpG和Poly(I:C)作为混合佐剂的纳米粒子激活的抗原提呈细胞制备的纳米疫苗治疗效果好于只使用一种CpG和Poly(I:C)混合佐剂的纳米粒子激活的抗原提呈细胞制备的纳米疫苗。综上所述,本发明所述的纳米疫苗对癌症具有良好的治疗效果。As shown in Figure 16, tumors in the PBS control group all grew rapidly. Compared with the control group, the tumor growth rate of the mice treated with the nanovaccine prepared from the nanoparticle-activated antigen-presenting cells was significantly slower, and the survival period was significantly prolonged. Moreover, the nanovaccine prepared by the nanoparticle-activated antigen-presenting cells with the addition of substances that increase lysosomal escape is better than the nanovaccine prepared by the antigen-presenting cells without the addition of the nanoparticle-activated lysosomal escape. Moreover, the nanovaccine prepared from nanoparticle-activated antigen-presenting cells using two CpG and Poly(I:C) adjuvants as mixed adjuvants was better than the one using only one CpG and Poly(I:C) mixed adjuvant. Nanovaccine prepared from nanoparticle-activated antigen-presenting cells. To sum up, the nano-vaccine of the present invention has a good therapeutic effect on cancer.
实施例16纳米疫苗用于乳腺癌的预防Example 16 Nano-vaccine for breast cancer prevention
本实施例以4T1小鼠三阴性乳腺癌为癌症模型来说明如何采用负载癌细胞全细胞抗原的微米粒子激活的抗原提呈细胞制备纳米疫苗用于预防乳腺癌。本实施例中,首先对乳腺癌细胞进行灭活和变性处理,尔后裂解细胞,并以辛基葡萄糖苷溶解裂解癌细胞中的非水溶性组分。然后,以PLGA为微米粒子骨架材料,以CpG2007(B类Toll样受体9激动剂)、CpG2216(A类Toll样受体9激动剂)、和Poly ICLC(Toll样受体3激动剂)为免疫佐剂,以聚精氨酸和聚赖氨酸为增强溶酶体逃逸的物质,制备负载有癌细胞全细胞组分的微米粒子,然后使用该微米粒子激活抗原提呈细胞,并制备基于被激活的抗原提呈细胞的纳米疫苗预防癌症。This example uses 4T1 mouse triple-negative breast cancer as a cancer model to illustrate how to use antigen-presenting cells activated by microparticles loaded with cancer cell whole cell antigens to prepare nano-vaccine for breast cancer prevention. In this example, breast cancer cells were first inactivated and denatured, and then the cells were lysed, and the water-insoluble components in the cancer cells were lysed and lysed with octyl glucoside. Then, PLGA was used as the microparticle framework material, and CpG2007 (Class B Toll-like receptor 9 agonist), CpG2216 (Class A Toll-like receptor 9 agonist), and Poly ICLC (Toll-
(1)癌细胞的裂解(1) Lysis of cancer cells
将培养的4T1细胞在400g离心5分钟,然后用PBS洗涤两遍后重悬于超纯水中。所得癌细胞分别采用紫外线和高温加热进行灭活和变性处理,然后加入超纯水并反复冻融5次辅以超声裂解癌细胞,将细胞裂解物在5000g离心10分钟,上清液即为水溶性组分,将沉淀物使用10%辛基葡萄糖苷溶解后即为溶解后的原非水溶性组分,将水溶性组分和非水溶性组分按质量比2:1混合,即为制备微米粒子所需的裂解物组分。The cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water. The obtained cancer cells were inactivated and denatured by ultraviolet and high temperature heating respectively, and then ultrapure water was added and freeze-thawed 5 times, supplemented by ultrasonic lysis, and the cell lysate was centrifuged at 5000g for 10 minutes, and the supernatant was water-soluble. It is the original water-insoluble component after dissolving the precipitate with 10% octyl glucoside, and the water-soluble component and the water-insoluble component are mixed in a mass ratio of 2:1, that is, the preparation Lysate components required for microparticles.
(2)微米粒子系统的制备(2) Preparation of microparticle system
本实施例中制备微米粒子系统及作为对照微米粒子采用复乳法,微米粒子骨架材料PLGA分子量为38KDa-54KDa,所采用的免疫佐剂为CpG2007(B类)、CpG2216(A类)和PolyICLC,所采用的溶酶体逃逸增加物质为聚精氨酸和聚赖氨酸。制备时先采用复乳法制备内部负载裂解物组分、佐剂和聚精氨酸和聚赖氨酸的微米粒子,然后将100mg微米粒子在9000g离心20分钟,使用10mL含4%海藻糖的超纯水重悬后干燥48h后备用。该微米粒子平均粒径为3.1μm左右,表面电位为-7mV左右;每1mg PLGA微米粒子约负载110μg蛋白质或多肽组分,含CpG2007(B类)、CpG 2216(A类)和Poly ICLC各0.01mg,含聚精氨酸和聚赖氨酸各0.02mg。对照微米粒子2制备材料和制备方法与如上方法相同,但是对照微米粒子2所负载的CpG为两种A类的CpG1585(A类)和CpG2216(A类),对照微米粒子2平均粒径为3.1μm左右,表面电位为-7mV左右;每1mg PLGA微米粒子约负载110μg蛋白质或多肽组分,含CpG1585(A类)、CpG2216(A类)和Poly ICLC各0.01mg,含聚精氨酸和聚赖氨酸各0.02mg。In this example, the microparticle system was prepared and used as a control microparticle by the double emulsion method. The molecular weight of the microparticle skeleton material PLGA was 38KDa-54KDa, and the immune adjuvants used were CpG2007 (type B), CpG2216 (type A) and PolyICLC, The lysosomal escape enhancing substances employed were polyarginine and polylysine. During preparation, the lysate component, adjuvant and microparticles loaded with polyarginine and polylysine were prepared by the double emulsion method, and then 100 mg of microparticles were centrifuged at 9000g for 20 minutes, and 10 mL of 4% trehalose was used. Resuspend in ultrapure water and dry for 48h before use. The average particle size of the microparticles is about 3.1μm, and the surface potential is about -7mV; each 1mg PLGA microparticle is loaded with about 110μg protein or polypeptide components, including CpG2007 (type B), CpG 2216 (type A) and Poly ICLC each 0.01 mg, containing 0.02 mg each of polyarginine and polylysine. The preparation material and preparation method of the
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用DC2.4细胞系作为抗原提呈细胞。This example uses the DC2.4 cell line as antigen presenting cells.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的微米粒子(1000μg)与DC2.4(1000万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD80抗体(10ng/mL)。Microparticles (1000 μg) loaded with whole cell components of cancer cells were co-incubated with DC2.4 (10 million) in 15 mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contained GM - CSF (2000 U/mL), IL-2 (500 U/mL), IL-7 (200 U/mL), IL-12 (200 U/mL) and CD80 antibody (10 ng/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,将上清液通过0.45μm的膜过滤挤出,然后在12000g离心90分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗,纳米疫苗粒径平均为250纳米。Incubated DCs were collected by centrifugation at 400 g for 5 min, cells were washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, cells were resuspended in PBS water and sonicated at 4°C at low power (20W) 2 minutes. The samples were then centrifuged at 3000g for 15 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 8000g for 15 minutes, the supernatant was extruded by filtration through a 0.45 μm membrane, and then collected after centrifugation at 12000g for 90 minutes The supernatant was discarded to collect the precipitate, and the precipitate was resuspended in PBS to obtain a nano-vaccine. The average particle size of the nano-vaccine was 250 nm.
(6)纳米疫苗癌症的预防(6) Nano-vaccine cancer prevention
选取6-8周的雌性BALB/c为模型小鼠制备乳腺癌荷瘤小鼠。在小鼠接种癌细胞前第-35天、第-21天和第-7天分别给每只小鼠皮下接种50μg纳米疫苗或者PBS。在第0天给每只小鼠皮下注射接种1×106个4T1细胞,从第3天开始每3天记录一次小鼠肿瘤体积的大小,小鼠肿瘤生长和生存期检测方法同上。Female BALB/c aged 6-8 weeks were selected as model mice to prepare breast cancer tumor-bearing mice. Each mouse was subcutaneously inoculated with 50 μg nanovaccine or PBS on days -35, -21 and -7 days before inoculation with cancer cells. On
(7)实验结果(7) Experimental results
如图17所示,与对照组相比,微米粒子激活的抗原提呈细胞制备的纳米疫苗处理的小鼠肿瘤生长速度明显变慢且小鼠生存期明显延长。而且,使用一种B类Toll样受体9激动剂与和一种A类Toll样受体9激动剂与Toll样受体3激动剂联用作为混合佐剂去激活抗原提呈细胞效果好于使用两种A类Toll样受体9激动剂与Toll样受体3激动剂联用作为混合佐剂去激活抗原提呈细胞。由此可见,本发明所述的基于被激活的抗原提呈细胞的纳米疫苗对乳腺癌具有预防效果。As shown in Figure 17, compared with the control group, the tumor growth rate of the mice treated with the nanovaccine prepared from the antigen-presenting cells activated by microparticles was significantly slower and the survival time of the mice was significantly prolonged. Moreover, the use of a class B Toll-like receptor 9 agonist in combination with a class A Toll-like receptor 9 agonist and a Toll-
实施例17纳米疫苗用于乳腺癌的预防Example 17 Nano-vaccine for breast cancer prevention
本实施例以4T1小鼠三阴性乳腺癌为癌症模型来说明如何微米粒子激活抗原提呈细胞后制备纳米疫苗用于预防癌症。This example uses 4T1 mouse triple-negative breast cancer as a cancer model to illustrate how to prepare nano-vaccine for cancer prevention after microparticles activate antigen-presenting cells.
(1)癌细胞的裂解(1) Lysis of cancer cells
将培养的4T1细胞在400g离心5分钟,然后用PBS洗涤两遍后重悬于超纯水中。所得癌细胞分别采用紫外线和高温加热进行灭活和变性处理,然后使用8M尿素水溶液(含500mM氯化钠)裂解癌细胞并溶解裂解物组分,即为制备微米粒子系统的抗原组分。The cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water. The obtained cancer cells were inactivated and denatured by ultraviolet rays and high temperature heating respectively, and then 8M urea aqueous solution (containing 500mM sodium chloride) was used to lyse the cancer cells and dissolve the lysate components, which are the antigen components of the microparticle system.
(2)微米粒子系统的制备(2) Preparation of microparticle system
本实施例中制备微米粒子系统及作为对照微米粒子采用复乳法,微米粒子骨架材料为未修饰的PLA和甘露糖修饰的PLA,分子量都为40KDa,未修饰的PLA和甘露糖修饰的PLA的比例为4:1。所采用的免疫佐剂为CpG2006、CpG2216和Poly ICLC,所采用的溶酶体逃逸增加物质为精氨酸和组氨酸。制备时先采用复乳法制备内部负载裂解物组分、佐剂、精氨酸和组氨酸的微米粒子,尔后,将100mg微米粒子在9000g离心20分钟,使用10mL含4%海藻糖的超纯水重悬后干燥48h后备用。该微米粒子系统平均粒径为2.1μm左右,微米粒子系统表面电位为-7mV左右;每1mg PLGA微米粒子约负载100μg蛋白质或多肽组分,含CpG2006,CpG2216和Poly ICLC各0.01mg,含精氨酸和组氨酸各0.05mg。对照微米粒2制备材料和制备方法与本实施例所述微米粒子相同,粒径为2.1μm左右,表面电位为-7mV左右,只负载精氨酸和组氨酸和等量的细胞裂解物组分,而不负载任何佐剂。In this example, the microparticle system was prepared and used as a control microparticle by the double emulsion method. The framework materials of the microparticles were unmodified PLA and mannose-modified PLA, and the molecular weights were both 40KDa. The unmodified PLA and mannose-modified PLA had The ratio is 4:1. The used immune adjuvants were CpG2006, CpG2216 and Poly ICLC, and the used lysosomal escape-increasing substances were arginine and histidine. During the preparation, the double emulsion method was used to prepare microparticles loaded with lysate components, adjuvants, arginine and histidine. Then, 100 mg of microparticles were centrifuged at 9000g for 20 minutes, and 10 mL of ultra-high-density aliquot containing 4% trehalose was used. Resuspend in pure water and dry for 48 hours before use. The average particle size of the microparticle system is about 2.1μm, and the surface potential of the microparticle system is about -7mV; each 1mg PLGA microparticle is loaded with about 100μg protein or polypeptide components, including 0.01mg each of CpG2006, CpG2216 and Poly ICLC, and arginine. 0.05 mg each of acid and histidine. The preparation material and preparation method of the
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC和B作为抗原提呈细胞。BMDC制备方法同实施例2。B细胞来自小鼠外周血PBMC,制备方法同上。This example used BMDC and B as antigen presenting cells. The preparation method of BMDC is the same as that in Example 2. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将微米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2);孵育体系中含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)和CD40抗体(20mg/mL)。Microparticles (1000 μg) were co-incubated with BMDC (5 million) and B cells (5 million) in 15 mL high glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contained GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500U/mL) and CD80 antibody (10ng/mL) and CD40 antibody (20 mg/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,然后在16000g离心60分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后通过0.22μm的膜过滤挤出,即得纳米疫苗,纳米疫苗粒径为150纳米。Incubated DCs and B cells were collected by centrifugation at 400 g for 5 min, then washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, resuspended in PBS water at 4°C low power ( 20W) sonicated for 2 minutes. The samples were then centrifuged at 3000g for 15 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 8000g for 15 minutes, and then after 60 minutes at 16000g. The supernatant was discarded to collect the pellet and the pellet was placed in PBS After resuspension, it is filtered and extruded through a 0.22 μm membrane to obtain a nano-vaccine, and the particle size of the nano-vaccine is 150 nm.
(6)癌细胞特异性T细胞用于癌症的预防(6) Cancer cell-specific T cells for cancer prevention
选取6-8周的雌性BALB/c为模型小鼠制备乳腺癌荷瘤小鼠。在小鼠接种癌细胞前第-35天、第-21天和第-7天分别每只小鼠接种50μg纳米疫苗或者PBS。同时在第0天给每只小鼠皮下注射接种1×106个4T1细胞,小鼠肿瘤体积以及生存期监测方法同上。Female BALB/c aged 6-8 weeks were selected as model mice to prepare breast cancer tumor-bearing mice. Each mouse was inoculated with 50 μg of nanovaccine or PBS on days -35, -21 and -7 days before inoculation of cancer cells. At the same time, 1×10 6 4T1 cells were subcutaneously injected into each mouse on the 0th day, and the methods for monitoring the tumor volume and survival period of the mice were the same as above.
(7)实验结果(7) Experimental results
如图18所示,与对照组相比,使用微米粒激活的抗原提呈细胞制备的纳米疫苗处理的小鼠,其肿瘤生长速度明显变慢且小鼠生存期明显延长。而且,含有增加溶酶体逃逸功能的物质和混合佐剂的微米粒子激活的抗原提呈细胞制备的纳米疫苗效果好于只含有溶酶体逃逸功能的物质而不含有混合佐剂的微米粒子激活的抗原提呈细胞制备的纳米疫苗。由此可见,本发明所述的基于抗原提呈细胞的纳米疫苗对乳腺癌具有预防效果,而且混合佐剂的使用有助于抗原提呈细胞的激活和后续纳米疫苗的活性。As shown in Figure 18, compared with the control group, the mice treated with the nanovaccine prepared by using microparticle-activated antigen-presenting cells had significantly slower tumor growth and significantly longer survival. Moreover, the nano-vaccine prepared by microparticle-activated antigen-presenting cells containing substances that increase lysosomal escape function and mixed adjuvants is better than that of microparticles activated by substances containing only lysosomal escape functions without mixed adjuvants. Nanovaccine prepared from antigen-presenting cells. It can be seen that the nano-vaccine based on antigen-presenting cells of the present invention has a preventive effect on breast cancer, and the use of mixed adjuvants is helpful for the activation of antigen-presenting cells and the subsequent activity of the nano-vaccine.
实施例18纳米疫苗用于癌症的治疗Example 18 Nano-vaccine for the treatment of cancer
本实施例以小鼠黑色素瘤为癌症模型来说明如何使用纳米粒子激活的抗原提呈细胞制备纳米疫苗用于治疗黑色素瘤。本实施例中,首先裂解肿瘤组织和癌细胞以制备水溶性组分,然后,以PLGA为骨架材料,以Poly(I:C)和CpG1018为免疫佐剂,以R8(RRRRRRRR)多肽为溶解溶酶体逃逸能力的物质,制备负载水溶性组分的纳米粒子,然后使用纳米粒子激活的抗原提呈细胞制备纳米疫苗治疗癌症。This example uses mouse melanoma as a cancer model to illustrate how to use nanoparticle-activated antigen-presenting cells to prepare a nanovaccine for the treatment of melanoma. In this example, tumor tissue and cancer cells were firstly lysed to prepare water-soluble components, then, PLGA was used as the skeleton material, Poly(I:C) and CpG1018 were used as immune adjuvants, and R8(RRRRRRRR) polypeptide was used as the lysing agent. Substances with the ability to escape from enzymes, prepare nanoparticles loaded with water-soluble components, and then use nanoparticle-activated antigen-presenting cells to prepare nanovaccine to treat cancer.
(1)肿瘤组织和癌细胞的裂解及各组分的收集(1) Lysis of tumor tissue and cancer cells and collection of components
收集肿瘤组织时先在每只C57BL/6小鼠背部皮下接种1.5×105个B16F10细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织,将肿瘤组织切块后研磨,通过细胞过滤网加入适量纯水并反复冻融5次(可伴有超声)以破坏裂解所得样品,加入核酸酶作用10分钟后在95℃加热10分钟灭活核酸酶;收集培养的B16F10癌细胞系时,先离心去除培养基后使用PBS洗涤两次并离心收集癌细胞,将癌细胞在超纯水中重悬,反复冻融3次,并伴有超声破坏裂解癌细胞,尔后在样品中加入核酸酶作用10分钟后在95℃加热5分钟灭活核酸酶。待肿瘤组织或癌细胞酶作用处理后,将裂解物以5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性组分。将肿瘤组织的水溶性组分和癌细胞的水溶性组分按质量比1:1混合,即为制备纳米粒子系统的抗原原料来源。When collecting tumor tissue, firstly inoculate 1.5×10 5 B16F10 cells subcutaneously on the back of each C57BL/ 6 mouse. When the tumor grows to a volume of about 1000 mm, the mice are sacrificed and the tumor tissue is excised. The tumor tissue is cut into pieces. Grind, add an appropriate amount of pure water through the cell strainer and freeze and thaw 5 times (accompanied by ultrasound) to destroy the lysed samples, add nuclease for 10 minutes, then heat at 95 °C for 10 minutes to inactivate the nuclease; collect the cultured B16F10 For cancer cell lines, the culture medium was first removed by centrifugation, then washed twice with PBS, and the cancer cells were collected by centrifugation. The cancer cells were resuspended in ultrapure water, freeze-thawed three times, and lysed cancer cells with ultrasonic damage. Nuclease was added to the sample for 10 minutes and then heated at 95°C for 5 minutes to inactivate the nuclease. After the tumor tissue or cancer cells were enzymatically treated, the lysate was centrifuged at 5,000 g for 5 minutes, and the supernatant was collected as a water-soluble component soluble in pure water. The water-soluble components of tumor tissue and the water-soluble components of cancer cells are mixed in a mass ratio of 1:1, which is the source of antigen raw materials for preparing the nanoparticle system.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒子采用复乳法制备。所采用的纳米粒子制备材料PLGA分子量为7KDa-17KDa,所采用的免疫佐剂为poly(I:C)和CpG1018,R8多肽为增加溶酶体逃逸的物质,且佐剂和R8多肽负载于纳米粒子内。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解液组分、佐剂和R8多肽,然后将100mg纳米粒子在12000g离心25分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h备用。该纳米粒子平均粒径为250nm左右,纳米粒子表面电位为-5mV左右;每1mg PLGA纳米粒子约负载120μg蛋白质或多肽组分,每1mg PLGA纳米粒所负载的poly(I:C)和CpG1018免疫佐剂各0.03mg,负载0.03mgR8多肽。In this example, the nanoparticles were prepared by the double emulsion method. The molecular weight of the nanoparticle preparation material PLGA is 7KDa-17KDa, the immunoadjuvant used is poly(I:C) and CpG1018, the R8 polypeptide is a substance that increases lysosome escape, and the adjuvant and R8 polypeptide are loaded on the nanoparticle. within the particles. The preparation method was as described above. In the preparation process, the lysate components, adjuvant and R8 polypeptide were first loaded into the nanoparticles by the double emulsion method, and then 100 mg of the nanoparticles were centrifuged at 12,000 g for 25 minutes, and 10 mL of 4% seaweed was used. The sugar was resuspended in ultrapure water and freeze-dried for 48 h for use. The average particle size of the nanoparticle is about 250nm, and the surface potential of the nanoparticle is about -5mV; each 1mg PLGA nanoparticle is loaded with about 120μg protein or polypeptide components, and each 1mg PLGA nanoparticle is loaded with poly(I:C) and CpG1018 immune Each adjuvant is 0.03 mg, and the R8 polypeptide is loaded with 0.03 mg.
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC和B作为抗原提呈细胞。BMDC制备方法同实施例2。B细胞来自小鼠外周血PBMC,制备方法同上。This example used BMDC and B as antigen presenting cells. The preparation method of BMDC is the same as that in Example 2. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将纳米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育24小时(37℃,5%CO2);或者将BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育24小时(37℃,5%CO2)。两种情况下孵育体系中都含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、和CD80抗体(10ng/mL)和CD40抗体(20mg/mL)。Nanoparticles (1000 μg) were co-incubated with BMDC (5 million) and B cells (5 million) in 15 mL high glucose DMEM complete medium for 24 hours (37°C, 5% CO 2 ); or BMDC (5 million) ) and B cells (5 million) were co-incubated in 15 mL of high glucose DMEM complete medium for 24 hours (37°C, 5% CO 2 ). In both cases the incubation system contained GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), and CD80 antibody (10ng /mL) and CD40 antibody (20mg/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在4000g离心5分钟并收集上清液,将上清液在6000g离心10分钟后收集上清液,将上清液通过0.22μm的膜过滤挤出,然后在17000g离心40分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得抗原提呈细胞细胞膜组分。将10mL抗原提呈细胞细胞膜组分(10mg)与50mg步骤(2)制备的纳米粒子混合,然后在4℃孵育15分钟后通过0.45μm的膜过滤挤出,然后在17000g离心40分钟后收集弃去上清液收集沉淀,将沉淀在含有冻干保护剂(2%海藻糖+2%甘露醇+赖氨酸)的水溶液中重悬后冷冻干燥48小时后即得纳米疫苗。使用被激活的混合抗原提呈细胞的细胞膜组分负载到纳米粒子表面制备的纳米疫苗为纳米疫苗1,粒径为260nm;使用未被激活的混合抗原提呈细胞细胞膜组分负载到纳米粒子表面制备的纳米疫苗为纳米疫苗2,粒径为260nm。Incubated DCs and B cells were collected by centrifugation at 400 g for 5 min, then washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, resuspended in PBS water at 4°C low power ( 20W) sonicated for 2 minutes. The samples were then centrifuged at 4000g for 5 minutes and the supernatant was collected, the supernatant was collected after centrifugation at 6000g for 10 minutes, the supernatant was extruded by filtration through a 0.22 μm membrane, and then collected after centrifugation at 17000g for 40 minutes Discard the supernatant to collect the pellet, and resuspend the pellet in PBS to obtain the cell membrane fraction of antigen-presenting cells. 10 mL of antigen-presenting cell membrane fraction (10 mg) was mixed with 50 mg of the nanoparticles prepared in step (2), then incubated at 4°C for 15 minutes, filtered and extruded through a 0.45 μm membrane, and then collected and discarded after centrifugation at 17,000 g for 40 minutes. The supernatant was removed to collect the precipitate, the precipitate was resuspended in an aqueous solution containing a lyoprotectant (2% trehalose + 2% mannitol + lysine), and the nanovaccine was obtained after lyophilization for 48 hours. The nanovaccine prepared by using the cell membrane components of the activated mixed antigen-presenting cells to be loaded on the surface of the nanoparticles is
(6)纳米疫苗治疗癌症(6) Nano-vaccine for cancer treatment
选取6-8周的雌性C57BL/6为模型小鼠制备黑色素瘤荷瘤小鼠。在第0天给每只小鼠背部右下方皮下接种1.5×105个B16F10细胞。在接种黑色素瘤后第4天、第7天、第10天、第15天和第20天分别皮下注射50μg纳米疫苗或者PBS。小鼠肿瘤生长和生存期监测方法同上。Select 6-8 week old female C57BL/6 as model mice to prepare melanoma tumor-bearing mice. 1.5 x 105 B16F10 cells were inoculated subcutaneously in the lower right back of each mouse on
(7)实验结果(7) Experimental results
如图19所示,与对照组相比,纳米疫苗处理的小鼠其肿瘤生长速度明显变慢且小鼠生存期明显延长。而且,使用纳米粒子激活的抗原提呈细胞制备的1纳米疫苗效果明显好于未经纳米粒子激活的抗原提呈细胞制备的纳米疫苗2。由此可见,本发明所述纳米粒子激活能够提高基于抗原提呈细胞的纳米疫苗的功效。As shown in Figure 19, compared with the control group, the tumor growth rate of the nanovaccine-treated mice was significantly slower and the survival time of the mice was significantly prolonged. Moreover, the effect of
实施例19纳米疫苗用于结肠癌的治疗Example 19 Nano-vaccine for the treatment of colon cancer
本实施例以小鼠结肠癌为癌症模型来说明如何使用负载有来源于结肠癌肿瘤组织的癌细胞全细胞抗原的纳米粒子激活的抗原提呈细胞制备纳米疫苗治疗结肠癌。本实施例中,首先使用8M尿素水溶液裂解结肠癌肿瘤组织并溶解裂解组分,然后,以PLGA为骨架材料,以Poly(I:C)、CpG2336和CpG2006为佐剂,以NH4HCO3为增加溶酶体逃逸物质,制备纳米粒子,然后使用纳米粒子激活的抗原提呈细胞制备纳米疫苗用于癌症治疗。This example uses mouse colon cancer as a cancer model to illustrate how to prepare a nanovaccine to treat colon cancer using nanoparticle-activated antigen-presenting cells loaded with cancer cell whole cell antigens derived from colon cancer tumor tissue. In this example, 8M urea aqueous solution was used to lyse colon cancer tumor tissue and the lysis components were firstly dissolved. Then, PLGA was used as the framework material, Poly(I:C), CpG2336 and CpG2006 were used as adjuvants, and NH 4 HCO 3 was used as adjuvant. Increase lysosomal escape substances, prepare nanoparticles, and then use nanoparticle-activated antigen-presenting cells to prepare nanovaccine for cancer treatment.
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
收集肿瘤组织时先在每只C57BL/6小鼠背部皮下接种2×106个MC38结肠癌细胞,在肿瘤长到体积分别为约1000mm3时处死小鼠并摘取肿瘤组织,将肿瘤组织切块后研磨,通过细胞过滤网加入8M尿素水溶液理解肿瘤组织并溶解裂解后全细胞组分。以上即为制备纳米粒子的抗原原料来源。When collecting tumor tissue, 2 × 10 6 MC38 colon cancer cells were subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of about 1000 mm, the mice were sacrificed and the tumor tissue was excised. After the blocks were ground, 8M aqueous urea solution was added through a cell strainer to dissociate the tumor tissue and lyse the whole cell fraction after lysis. The above is the source of antigenic raw materials for the preparation of nanoparticles.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒子采用复乳法制备。纳米粒子1的制备材料PLGA分子量为7KDa-17KDa,以Poly(I:C)和CpG为佐剂,以NH4HCO3为增加溶酶体逃逸物质,且佐剂和NH4HCO3负载于纳米粒子内;制备方法如前所述,在制备过程中首先在纳米粒子内部负载裂解液组分和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h后备用;该纳米粒子平均粒径为260nm左右,表面电位为-7mV左右;每1mgPLGA纳米粒子约负载90μg蛋白质和多肽组分,每1mg PLGA纳米粒所负载的poly(I:C)、CpG2336和CpG2006免疫佐剂各0.02mg,负载NH4HCO3 0.01mg。纳米粒子2的制备材料和制备方法同纳米粒子1,粒径为260nm左右,表面电位为-7mV左右,每1mg PLGA纳米粒子约负载90μg蛋白质和多肽组分,每1mg PLGA纳米粒负载NH4HCO3 0.01mg,负载CpG2336和CpG2006各0.03mg。In this example, the nanoparticles were prepared by the double emulsion method. The preparation material of
(3)抗原提呈细胞的制备(3) Preparation of antigen presenting cells
本实施例使用BMDC和B作为抗原提呈细胞。BMDC制备方法同实施例2。B细胞来自小鼠外周血PBMC,制备方法同上。This example used BMDC and B as antigen presenting cells. The preparation method of BMDC is the same as that in Example 2. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen presenting cells
将负载癌细胞全细胞组分的纳米粒子1(500μg)或纳米粒2(500μg)与BMDC(250万个)及B细胞(250万个)在10mL高糖DMEM完全培养基中共孵育69小时(37℃,5%CO2),孵育体系中含有GM-CSF(500U/mL)、IL-2(300U/mL)、IL-7(200U/mL)、IL-12(500U/mL)和CD80抗体(10ng/mL)和CD40抗体(20mg/mL);或者将负载癌细胞全细胞组分的纳米粒子1(500μg)与BMDC(250万个)及B细胞(250万个)在10mL高糖DMEM完全培养基中共孵育69小时(37℃,5%CO2),孵育体系中含有GM-CSF(500U/mL)、IL-12(1000U/mL)和CD80抗体(10ng/mL)。Nanoparticles 1 (500 μg) or nanoparticle 2 (500 μg) loaded with cancer cell whole cell fractions were co-incubated with BMDC (2.5 million) and B cells (2.5 million) in 10 mL of high-glucose DMEM complete medium for 69 hours ( 37°C, 5% CO 2 ), the incubation system contains GM-CSF (500U/mL), IL-2 (300U/mL), IL-7 (200U/mL), IL-12 (500U/mL) and CD80 Antibody (10ng/mL) and CD40 antibody (20mg/mL); or nanoparticle 1 (500μg) loaded with whole cell fraction of cancer cells with BMDC (2.5 million) and B cells (2.5 million) in 10 mL of high glucose DMEM complete medium was incubated for 69 hours (37°C, 5% CO2) containing GM-CSF (500U/mL), IL-12 (1000U/mL) and CD80 antibody (10ng/mL).
(5)抗原提呈细胞来源的纳米疫苗的制备(5) Preparation of antigen-presenting cell-derived nanovaccine
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心30分钟弃去沉淀只收集上清液,将上清液在8000g离心15分钟后弃去沉淀收集上清液,将上清液通过0.22μm的膜过滤挤出,然后在18000g离心90分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米疫苗,纳米疫苗粒径为120纳米。Incubated DCs and B cells were collected by centrifugation at 400 g for 5 min, then washed twice with 4°C phosphate buffered saline (PBS) containing protease inhibitors, resuspended in PBS water at 4°C low power ( 20W) sonicated for 2 minutes. The sample was then centrifuged at 3000g for 30 minutes to discard the precipitate and only the supernatant was collected. The supernatant was centrifuged at 8000g for 15 minutes and the precipitate was discarded to collect the supernatant. The supernatant was filtered and extruded through a 0.22 μm membrane, and then After centrifugation at 18000g for 90 minutes, the supernatant was discarded and the precipitate was collected, and the precipitate was resuspended in PBS to obtain a nano-vaccine, and the particle size of the nano-vaccine was 120 nm.
(6)癌细胞特异性T细胞用于治疗癌症(6) Cancer cell-specific T cells for the treatment of cancer
选取6-8周的雌性C57BL/6为模型小鼠制备结肠癌小鼠。在第0天给每只小鼠背部右下方皮下接种2×106个MC38细胞。在接种结肠癌细胞后第6天、第9天、第12天、第15天、第20天和第25天分别给每只小鼠皮下注射50μg纳米疫苗。小鼠肿瘤生长和生存期监测方法同上。Colon cancer mice were prepared by selecting 6-8 week old female C57BL/6 as model mice. On
(7)实验结果(7) Experimental results
如图20所示,与对照组相比,纳米粒子激活的抗原提呈细胞制备的纳米疫苗处理小鼠后其肿瘤生长速度明显变慢且小鼠生存期明显延长。而且,使用同时负载裂解物组分、混合佐剂及溶酶体逃逸物质的纳米粒子激活的抗原提呈细胞制备的纳米疫苗明显好于使用同时负载裂解物组分、两种CpG佐剂和溶酶体逃逸物质的纳米粒子激活的抗原提呈细胞制备的纳米疫苗。而且,在纳米粒子激活抗原提呈细胞过程中加入IL-7、IL-2和抗体共孵育的效果好于不加入上述两种白介素和抗体共孵育。由此可见,本发明所述的基于纳米粒子激活的抗原提呈细胞制备的纳米疫苗对癌症具有优异治疗效果,而混合佐剂的使用以及激活抗原提呈细胞时加入抗体和IL-7、IL-2对最终制备的纳米疫苗有增强效果。As shown in Figure 20, compared with the control group, the tumor growth rate of the mice treated with the nanovaccine prepared by the nanoparticle-activated antigen-presenting cells was significantly slower and the survival time of the mice was significantly prolonged. Moreover, nanovaccine prepared using nanoparticle-activated antigen-presenting cells loaded with lysate fraction, mixed adjuvant, and lysosomal escape material was significantly better than that prepared with lysate fraction, two CpG adjuvants, and lysosomal escape materials. Nanovaccine prepared from antigen-presenting cells activated by nanoparticles of enzymatic escape substances. Moreover, the co-incubation effect of adding IL-7, IL-2 and antibody in the process of activating antigen-presenting cells by nanoparticles is better than that without adding the two interleukins and antibody. It can be seen that the nanovaccine prepared based on the nanoparticle-activated antigen-presenting cells of the present invention has an excellent therapeutic effect on cancer, and the use of mixed adjuvants and the addition of antibodies and IL-7, IL to activate the antigen-presenting cells -2 has an enhanced effect on the final prepared nano-vaccine.
显然,上述实施例仅仅是为清楚地说明所作的举例,并非对实施方式的限定。对于所属领域的普通技术人员来说,在上述说明的基础上还可以做出其它不同形式变化或变动。这里无需也无法对所有的实施方式予以穷举。而由此所引申出的显而易见的变化或变动仍处于本发明创造的保护范围之中。Obviously, the above-mentioned embodiments are only examples for clear description, and are not intended to limit the implementation manner. For those of ordinary skill in the art, other different forms of changes or modifications can also be made on the basis of the above description. There is no need and cannot be exhaustive of all implementations here. However, the obvious changes or changes derived from this are still within the protection scope of the present invention.
Claims (10)
Priority Applications (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210630310.6A CN114931632A (en) | 2022-06-06 | 2022-06-06 | Cancer vaccine based on antigen presenting cell membrane component and preparation method and application thereof |
PCT/CN2022/108960 WO2023236330A1 (en) | 2022-06-06 | 2022-07-29 | Cancer vaccine based on antigen-presenting cell membrane components, method for preparing same, and use thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210630310.6A CN114931632A (en) | 2022-06-06 | 2022-06-06 | Cancer vaccine based on antigen presenting cell membrane component and preparation method and application thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN114931632A true CN114931632A (en) | 2022-08-23 |
Family
ID=82866513
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202210630310.6A Pending CN114931632A (en) | 2022-06-06 | 2022-06-06 | Cancer vaccine based on antigen presenting cell membrane component and preparation method and application thereof |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN114931632A (en) |
WO (1) | WO2023236330A1 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN115554315A (en) * | 2022-07-01 | 2023-01-03 | 苏州尔生生物医药有限公司 | Cancer cell specific T cell vaccine and method for activating cancer cell specific T cells |
WO2024250376A1 (en) * | 2023-06-07 | 2024-12-12 | 苏州尔生生物医药有限公司 | In-vitro activated mixed cell cancer vaccine containing dendritic cells and b cells, and use thereof |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN119838004B (en) * | 2025-03-21 | 2025-07-18 | 中国医学科学院生物医学工程研究所 | A tumor immunotherapy nanovaccine encapsulated by engineered cell-derived vesicles and its application |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2021077566A1 (en) * | 2019-10-23 | 2021-04-29 | 北京大学 | Application of divalent manganese in preparing immunity enhancing drug or antitumor drug |
CN113041342A (en) * | 2021-03-24 | 2021-06-29 | 深圳先进技术研究院 | Nano artificial antigen presenting cell and preparation method and application thereof |
CN113440605A (en) * | 2020-03-26 | 2021-09-28 | 苏州大学 | Conveying system for whole cell components and application thereof |
CN114099655A (en) * | 2021-11-16 | 2022-03-01 | 苏州大学 | A vaccine system for preventing or treating cancer and its application |
CN114404580A (en) * | 2021-12-24 | 2022-04-29 | 苏州大学 | Dendritic cell cancer vaccine and application thereof |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR20140106389A (en) * | 2011-05-17 | 2014-09-03 | 솔리제닉스, 인크. | Thermostable vaccine compositions and methods of preparing same |
CN110090298A (en) * | 2019-05-09 | 2019-08-06 | 武汉大学 | A kind of cell membrane tumor vaccine and preparation method and application |
CN112245574B (en) * | 2020-10-23 | 2023-07-14 | 苏州尔生生物医药有限公司 | A targeted delivery system loaded with whole cell components and its application |
-
2022
- 2022-06-06 CN CN202210630310.6A patent/CN114931632A/en active Pending
- 2022-07-29 WO PCT/CN2022/108960 patent/WO2023236330A1/en active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2021077566A1 (en) * | 2019-10-23 | 2021-04-29 | 北京大学 | Application of divalent manganese in preparing immunity enhancing drug or antitumor drug |
CN113440605A (en) * | 2020-03-26 | 2021-09-28 | 苏州大学 | Conveying system for whole cell components and application thereof |
CN113041342A (en) * | 2021-03-24 | 2021-06-29 | 深圳先进技术研究院 | Nano artificial antigen presenting cell and preparation method and application thereof |
CN114099655A (en) * | 2021-11-16 | 2022-03-01 | 苏州大学 | A vaccine system for preventing or treating cancer and its application |
CN114404580A (en) * | 2021-12-24 | 2022-04-29 | 苏州大学 | Dendritic cell cancer vaccine and application thereof |
Non-Patent Citations (1)
Title |
---|
高俊潇;: "纳米颗粒疫苗在肿瘤免疫治疗中的研究进展", 化学与生物工程, no. 08, 14 August 2020 (2020-08-14), pages 1 - 3 * |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN115554315A (en) * | 2022-07-01 | 2023-01-03 | 苏州尔生生物医药有限公司 | Cancer cell specific T cell vaccine and method for activating cancer cell specific T cells |
WO2024000725A1 (en) * | 2022-07-01 | 2024-01-04 | 苏州尔生生物医药有限公司 | Cancer cell-specific t cell vaccine and method for activating cancer cell-specific t cells |
WO2024250376A1 (en) * | 2023-06-07 | 2024-12-12 | 苏州尔生生物医药有限公司 | In-vitro activated mixed cell cancer vaccine containing dendritic cells and b cells, and use thereof |
Also Published As
Publication number | Publication date |
---|---|
WO2023236330A1 (en) | 2023-12-14 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20250041414A1 (en) | Dendritic cell cancer vaccine and application thereof | |
CN114931632A (en) | Cancer vaccine based on antigen presenting cell membrane component and preparation method and application thereof | |
CN114099655A (en) | A vaccine system for preventing or treating cancer and its application | |
CN114225021A (en) | Vaccine system for preventing or treating cancer based on one or more cancer cell and/or tumor tissue whole cell components or mixture thereof | |
CN114288397B (en) | Vaccine system for preventing or treating cancer based on whole cell components of various cancer cells and/or tumor tissues, preparation and application thereof | |
CN113440605A (en) | Conveying system for whole cell components and application thereof | |
CN114931633A (en) | Preparation method and application of autoimmune disease vaccine derived from pre-activated antigen presenting cells | |
EP4534652A1 (en) | Specific t cell for preventing or treating cancer, and preparation method therefor | |
WO2024016688A1 (en) | Nucleic acid delivery particle based on activated antigen-presenting cells, nucleic acid delivery system and preparation method | |
CN114887049A (en) | Immunologic adjuvant composition, cancer vaccine based on composition and application of immunologic adjuvant composition | |
CN114288398A (en) | Application of cancer vaccine system based on whole cell components in the preparation of cross-prevention or treatment of heterologous cancer drugs | |
WO2024000724A1 (en) | Preparation method for vaccine loaded with cancer cell whole-cell component and mixed membrane component and use thereof | |
WO2023201787A1 (en) | Cancer-specific t cell-based cell system, lymphocyte drug and use thereof | |
WO2024250371A1 (en) | Cancer vaccine based on lysate components and synthetic cancer antigens in cancer cells and/or tumor tissues and preparation method | |
WO2024007388A1 (en) | Method and kit for detecting cancer cell-specific t cell | |
CN114958739A (en) | Cell system, application thereof and method for activating broad-spectrum cancer cell specific T cells | |
WO2024212380A1 (en) | Cancer vaccine based on part of components of cancer cells or tumor tissue, and preparation method therefor | |
WO2024250376A1 (en) | In-vitro activated mixed cell cancer vaccine containing dendritic cells and b cells, and use thereof | |
CN117122678A (en) | Method for preparing antigen delivery particles | |
WO2023082454A1 (en) | Vaccine system for preventing or treating diseases on basis of whole-cell components of one or more bacteria, and preparation method therefor and use thereof | |
CN115554315A (en) | Cancer cell specific T cell vaccine and method for activating cancer cell specific T cells | |
WO2025113111A1 (en) | Method for extracting antigen component, and delivery particle containing same and use | |
WO2025113109A1 (en) | Preparation method for antigen delivery particle and use thereof | |
CN117618602A (en) | Terminal sterilization method of cancer particle vaccine loaded with cancer cell lysate component and application of terminal sterilization method | |
WO2025059964A1 (en) | Method for preparing antigen-delivery particle |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |