CN108379569A - The DC vaccines of efficient load tumour antigen and its method of induced amplification specific for tumour antigen CTL - Google Patents

The DC vaccines of efficient load tumour antigen and its method of induced amplification specific for tumour antigen CTL Download PDF

Info

Publication number
CN108379569A
CN108379569A CN201810448476.XA CN201810448476A CN108379569A CN 108379569 A CN108379569 A CN 108379569A CN 201810448476 A CN201810448476 A CN 201810448476A CN 108379569 A CN108379569 A CN 108379569A
Authority
CN
China
Prior art keywords
cell
tumour
ctl
antigen
vaccines
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.)
Granted
Application number
CN201810448476.XA
Other languages
Chinese (zh)
Other versions
CN108379569B (en
Inventor
徐迎新
李力
梁凯
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beijing Bai Yining Medical Science And Technology Co Ltd
Original Assignee
Beijing Bai Yining Medical Science And Technology Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beijing Bai Yining Medical Science And Technology Co Ltd filed Critical Beijing Bai Yining Medical Science And Technology Co Ltd
Priority to CN201810448476.XA priority Critical patent/CN108379569B/en
Publication of CN108379569A publication Critical patent/CN108379569A/en
Application granted granted Critical
Publication of CN108379569B publication Critical patent/CN108379569B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0639Dendritic cells, e.g. Langherhans cells in the epidermis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/51B7 molecules, e.g. CD80, CD86, CD28 (ligand), CD152 (ligand)

Abstract

A kind of method that the present invention provides the DC vaccines and its induced amplification specific for tumour antigen CTL of efficient load tumour antigen.The DC vaccines are prepared by the following method to obtain:(1) tumour antigen is prepared;(2) it acquires, separation and high-efficient culture expand DC cells;(3) tumour antigen and activator sensitization is used to activate DC cells;(4) closing of DC cell surfaces PD L1 molecules.The present invention also provides sequential blood drawings in the method for the external evoked amplification CTL of DC vaccines, and the closing to external evoked 1 molecules of amplification CTL surface PD.The present invention solves the problems, such as that existing tumour antigen immunogenicity is not strong, Antigenic Target is not complete, patient tumour antigen is not easy to obtain, DC and CTL amplification in vitro lazy weights.DC vaccines can effectively induce specific for tumour antigen CTL in vitro and in vivo, and efficiently can generate specific killing to tumour, no obvious toxic-side effects, and potential applicability in clinical practice is wide.

Description

The DC vaccines and its induced amplification specific for tumour antigen of efficient load tumour antigen The method of CTL
Technical field
The present invention relates to medicine and biotechnology, specifically, it is thin to be related to a kind of low single core of initial amount peripheral blood Born of the same parents efficiently prepare self load or the DC vaccines of other source tumour antigens and its system of induced amplification specific for tumour antigen CTL Preparation Method.
Background technology
Tumour is the inevitable outcome of human evolution, and threatens the major disease of human health and life.China is swollen at present Tumor incidence rises year by year, and traditional operation, chemotherapy, radiotherapy etc. all has larger limitation.Operation can only cut off naked eyes Finding lump can not thoroughly remove tumour cell, the more vivo environment without changing tumor patient loss of equilibrium;Chemotherapy, radiotherapy can With fragmentation unit tumour cell, but quite a few entity tumor is insensitive to Radiotherapy chemotherapy, and many side effects are exempted to patient's Epidemic disease system and quality of life bring series of negative influence, can not also prevent the relapse and metastasis of tumour;Existing molecular targeted medicine Object is of limited application, and easily generates drug resistance.Therefore, there is an urgent need for open up new therapy approach and means.
Numerous studies show that recovery/reconstruction patients itself antineoplastic immune function is the indispensable strategy of oncotherapy One of.Immunotherapy of tumors has become the fourth-largest means of oncotherapy in recent years, and tumor-specific immunity treatment is weight In it is weight, influence tumor-specific immunity treatment several elements include:1) correctly can induce and expand sufficient amount has The Dendritic Cells (DC) and tumor specific cytotoxicity T cell (CTL) of powerful antigen presentation function;2) best tumour is anti- It is former;3) in the infiltration to tumor tissues of chemotactic tumor specific cytotoxicity T cell;4) the immunosupress machine of tumor microenvironment is cut off System.
The sources DC can be different, but are most easily to obtain mononuclearcell from peripheral blood separation.Since DC and CTL is expanded Relatively difficult, for the cell quantity for meeting needed for treatment, people usually take measures on the cell quantity of culture starting, wrap It includes:1) 2-3 days injection GM-CSF carry out bone marrow mobilization before mononuclearcell acquisition, to increase the ratio of peripheral mononuclear blood cell And absolute counting;2) blood cell separator is used, largely mononuclearcell is acquired by the thousands of milliliters of Peripheral Circulation.In this way Processing strong external intervention actually has been carried out to the marrow of patient and peripheral blood, especially for tumor patient art Afterwards, the organismic internal environments such as immune system disorderly after Radiotherapy chemotherapy have further resulted in very big interference.Therefore, clinical anxious A kind of body need to be had not significant impact, started with smaller cell culture initial amount, lived by culture amplification and antigen sensibilization After change, it can meet the DC vaccines of Treatment need and its effector cell CTL of induced amplification in cell quantity and phenotype.
Other than cell quantity, best tumour antigen is the key that ensure tumor-specific immunity treatment accuracy, It needs to meet three aspects:1) specificity and stronger immunogenicity of tumour antigen;2) multiple target point;3) individuation.It is so far Only, it has been found that some are in normal structure low expression and in the tumor associated antigen (TAA) of tumor tissues height expression;Minority exists Normal structure is not expressed (except testis and placenta), the tumour specific antigen (TSA) only expressed in tumor tissues;Pass through group Knit the tumour-specific polypeptides of sample storehouse and organization chip and the extensive screening of monoclonal antibody;Believed based on gene sequencing and biology Cease the neoantigen that credit analysis obtains (for the Neo-Antigen of individuation);But it for most of tumours, does not still obtain There is extensive representative tumour specific antigen recognized by many.Tumour antigen at present in existing tumor vaccine is each has something to recommend him: Full tumour cell tumor seedling and Tumor lysate tumor seedling antigen broad covered area, immunogenicity is strong, but more containing impurity, poor specificity; Tumor relative polypeptide high specificity, but less immunogenic;Anti-idiotype Antibody Vaccine is often single target spot;Genetic modification swells Tumor vaccine attempts to improve the immunogenicity of tumour or the activity of antigen presenting cell, but is generally also single target spot, and transgenosis is grasped Work itself is exactly a kind of damage to antigen presenting cell.Tumour cell discharge vesica antigen (excretion body) can meet multiple target point and The strong requirement of particulate antigen immunogenicity, but the antigen in cells and supernatant is only collected in preparation process, it has but lost swollen A large amount of antigens on oncocyte surface.Change in view of the polygenes of tumour cell, the height heterogeneity and tumour of tumor tissues are suffered from The height of person is personalized, and the specific active immunotherapy of tumour is made to still suffer from great challenge.In addition, tumor vaccine is as a kind of Biological agent is also contemplated that the efficiency of the obtained tumour antigen of production process from production angle.There is an urgent need to develop target spot covering surfaces Extensively (multiple target point), specificity is high, the strong tumour antigen preparation method of immunogenicity, to meet the novel DC vaccine antigens of individuation Load, and can strong induction in vitro and in vivo and amplification multiple target point tumor specific cytotoxicity T cell.
Tumor patient usually has different degrees of immune tolerance, and how break immune is resistant to, and repairs or excitation dendron shape is thin The powerful antigen submission ability of born of the same parents (DC), it is tumour immunity to make its abundant induced activation tumor specific cytotoxicity T cell (CTL) The key for the treatment of.Recently, effect of the immune detection point in oncotherapy receives more and more attention, PD-1 antibody and PD- L1 antibody is currently used to the treatment of tumour.But that people's common concern is the PD-1 that T cell surface is closed with PD-1 antibody Molecule, with the PD-L1 of PD-L1 antibody blocking tumor cells expressions, to activate or enhance the function of T cell.Actually not only swell Oncocyte can express PD-L1, and DC can similarly express PD-L1.The ripe DC being especially activated after load antigen can be with Height expression PD-L1, this is also one kind " brake " mechanism of DC inducing specific immunities, and unclamps this " brake " and effectively answer In the treatment of clinical disease, using the PD-L1 receptors of closing DC cell surfaces, realize that the antigen of DC is passed to the maximum extent It is in and activates the ability of CTL, the PD-1 molecules on the surfaces joint closing CTL, the tumor specific cytotoxicity for improving CTL is horizontal, right It is a kind of approach of worth exploration to break DC tolerances and the immunization therapy of tumour-specific, individuation.
Invention content
The object of the present invention is to provide a kind of DC vaccines of efficient load tumour antigen and its induced amplification tumour antigen are special The method of anisotropic CTL, the DC vaccines are using low initial amount peripheral blood mononuclear cells come efficiently to prepare load self or other Source tumour antigen effectively induces, extensive amplification multiple target point tumor specific cytotoxicity T cell in vitro and in vivo (CTL), it is used for the clinical treatment of tumor patient.
In order to realize the object of the invention, present invention firstly provides a kind of preparations of the DC vaccines of efficient load tumour antigen Method includes the following steps:
(1) tumour antigen is prepared;
(2) acquisition peripheral blood detaches mononuclearcell, with adherent low initial amount cell directional induction and amplification DC cells;
(3) tumour antigen and activator sensitization is used to activate DC cells;
(4) DC cell surface PD-L1 molecules are closed, DC vaccines are harvested.
In above-mentioned preparation method, tumour antigen and the peripheral blood of step (2) of step (1) come from same individual or different Body depends on clinical acquisition autologous tumor tissue or the feasibility of cell and the type of tumour antigen.
Step (1) the tumour antigen type is selected from Tumor Tissue Lysates, tumor cell lysate, tumor cell surface Proteantigen, tumour cell Membrane protein antigen, tumour cell excretion body, other tumours correlation or specific proteins, tomour specific Property polypeptide, be based on gene sequencing and bioinformatic analysis, predict and prepare one kind or more in the personalized neoantigen of height Kind.
The tumour antigen both can be used alone, and can also be used in mixed way.The implementation of the preferred version of the present invention In example, tumour antigen used is that tumor cell surface proteantigen, tumour cell Membrane protein antigen and tumour cell excretion body are pressed The mixture of mass ratio 1: 1: 2.
Step (1) tumour antigen is obtained by being chemically treated with the following methods of the sequential progress of physical heating:To swollen After arsenic trioxide culture is added in oncocyte, after tumour cell diminution is rounded, adherent rate reduces, after cytoplasm blebbing, 40-46 DEG C culture cell, to induce the expression of heat shock protein.
Preferably, to it is being detached from patient's tumor tissues or build be culture tumour cell in final concentration of 1umol/ is added 37 DEG C of the arsenic trioxide of L-15umol/L, 5%CO2Culture 4-24 hour, wait for tumour cell diminution be rounded, adherent rate reduction, After cytoplasm blebbing, 43 DEG C, 5%CO2Culture 1-12 hours.
In the preparation method of above-mentioned DC vaccines provided by the invention, step (2) acquires and the method that is separately cultured DC cells It is as follows:
1) separation of peripheral blood mononuclear cells:Anticoagulation is extracted from peripheral vein to be added on lymphocyte separation medium, from The heart detaches mononuclearcell, serum-free DC cell culture fluids is added, cell is resuspended, until cell concentration is 3 × 106-1.5×107A/ ml;
2) separation, directional induction and amplification of DC cells:Mononuclearcell adhere-wall culture 2-6 hours;It is sucked out not adherent thin Born of the same parents, gained attached cell are DC precursors;The rh-IL-4 and final concentration of final concentration 1000IU are added into DC precursors The th-GM-CSF of 1000IU, directional induction and amplification DC precursors 7-9 days, gained immature DC cell is for sensitization activation.
The present invention prepares DC vaccine approach key point in step (2) and is, preparing the DC vaccines per person-portion only needs The peripheries 40-50ml anticoagulation is acquired, with a large amount of DC of mononuclearcell induced amplification of low starting quantity.
One embodiment of the present of invention the step of in (2), the method for acquiring and being separately cultured DC cells is as follows:
1) separation of peripheral blood mononuclear cells:Extract periphery anticoagulation 40-50ml (preferably 45m1), Conventional density gradients Centrifugal process obtains mononuclearcell, serum-free DC cell culture fluids is added after washing, cell is resuspended, and carries out cell count, and adjustment is thin Born of the same parents a concentration of 3 × 106-1.5×107A/ml.
The serum-free DC cell culture mediums are special culture media (BYN-PD701), and the lymphocyte separation medium can come From commercially available commercially produced product, it is preferably purchased from Sigma Products.
2) separation, directional induction and amplification of DC cells:Cell is at 37 DEG C, 5%CO2In incubator, with serum-free DC cells Culture medium (BYN-PD701) adhere-wall culture 2-6 hours;The cell that non-attached cell can be used for the DC activation of tumour antigen sensitization is sucked out Malicious T cell (DC-AT) or cytokine induced kill cell (CIK) amplification cultivation, gained attached cell are that DC precursors are thin Born of the same parents;The rh-IL-4 and final concentration 1000IU of DC cells special culture medias, final concentration 1000IU is added into DC precursors Rh-GM-CSF, in 37 DEG C of 5%CO2In incubator, directional induction and amplification DC precursors 7-9 days, gained immature DC cell has Wait for that sensitization activates.According to cell growth and amplification situation, individuation timely and appropriate discovery adds the rh-IL-4 containing final concentration 1000IU With the serum-free DC cells special culture media (BYN-PD701) of the rh-GM-FCS of final concentration 1000IU.
In the preparation method of the DC vaccines of efficient load tumour antigen provided by the invention, it is thin that step (3) sensitization activates DC The method of born of the same parents is:Using the antigen in the autologous tumor source from peripheral blood donor, or according to the pathological examination of patient as a result, plus Enter the mixed rumour antigen of the corresponding tumour to match to pathological examination result, while activator is added, incubation obtains sensitization work The DC cells of change;The activator is bacterial product, complex immunity globulin, monoclonal antibody, micromolecular compound.
Preferably, the activator is Polyinosinic injection, anti-human CD 40 monoclonal antibody, TNF-d or bacterial product come The marketed products (injection) in source.In the embodiment of the present invention, the mixed rumour antigen of selection contains tumor cell surface antigen, Membrane antigen, tumour cell excretion body.It will be appreciated by those skilled in the art that mixed rumour antigen includes but not limited to following Any two or multiple mixing of tumour antigen:Tumor Tissue Lysates, tumor cell lysate, tumor cell surface albumen are anti- Original, tumour cell Membrane protein antigen, tumour cell excretion body, other tumours be related or specific proteins, tumour-specific polypeptides, Height personalization neoantigen based on gene sequencing.
Specifically, the method for step (3) tumour antigen and activator sensitization activation DC cells is as follows:To DC cell concentrations It is 5 × 106-1.5×107The mixed rumour antigen of 1/10 volume is added in the serum-free DC cell culture fluids of a/ml, and (tumour is thin Cellular surface proteantigen: tumour cell Membrane protein antigen: tumour cell excretion body mass ratio 1: 1: 2), while final concentration is added The activator of the Polyinosinic injection and final concentration 2-10 μ g/ml of 0.2-0.8 μ g/ml is incubated overnight, and it is thin to obtain ripe DC Born of the same parents.
It is highly preferred that the activator is selected from anti-human CD 40 monoclonal antibody.
In the preparation method of the DC vaccines of efficient load tumour antigen provided by the invention, step (4) is activated to sensitization Anti human PD-L 1 antibody is added in DC cells afterwards, is incubated 0.5-2 hours at 30-40 DEG C, to close PD-L1 points of DC cell surfaces Son.
Specifically, in step (4), the anti human PD-L 1 antibody of final concentration 1-10ug/ml is added into ripe DC cells, 37 DEG C of 5%CO2Under the conditions of be incubated 0.5-2 hours, close DC cell surface PD-L1 antigen molecules, harvest closing after DC cells, The DC vaccines of as efficient load tumour antigen, can be directly used for active immunity treatment, i.e., induced amplification CTL in vivo.
On the other hand, the present invention also provides obtained efficient load tumour antigen DC vaccines, induced amplification is swollen in vitro Purposes in tumor antigen specific CTL.The purposes includes two committed steps:(1) sequential to extract the non-patch of a small amount of peripheral blood separation Wall mononuclearcell, it is external evoked with DC vaccines, and priority primary culture medium and amplification culture medium culture expand CTL;(2) body The closing of the surfaces the CTL PD-1 molecules of outer induced amplification.It is specific as follows:
(1) tumour antigen sensitization activates the external evoked amplification tumor-specific CTLs of DC
Tumour antigen sensitization activation DC cell surface height expression CD80, CD83, CD86, HLA-DR (figure prepared by this method 1A, Figure 1B), illustrate that its maturity is higher, also can inducing tumor-specific CTL in vitro, therefore prepared in first time blood drawing swollen On the tumor antigen sensitization DC harvest same day, 40~50mL of same tumor patient anticoagulation (i.e. sequential blood drawing) is extracted for the second time, separation is single Not adherent mononuclearcell is sucked out in a nucleus adhere-wall culture, and DC is activated with 3-7: 1 with the tumour antigen sensitization of same day harvest Ratio mixes, and sets and CTL serum-frees primary culture medium (BYN-PT331) is added in culture bottle, 37 DEG C, 5%CO2Incubator culture, it is secondary Day anti-human CD3 monoclonal antibodies, anti-human CD28 monoclonal antibodies, rh-IL-2 is added to continue to cultivate.Then a according to cell growth and amplification situation Body timely and appropriate discovery adds the serum-free amplification culture medium (BYN-PT332) containing rh-IL-2 and carries out CTL amplification cultivations.
(2) closing of the surfaces CTL PD-1 molecules is harvested with CTL
CTL was cultivated to 11-14 days, and when cell log summit of growth, before CTL harvests, anti-human PD-1 Dan Ke are added Grand antibody 1-10ug/ml (final concentration), the PD-1 molecules on the surfaces closing CTL, 37 DEG C, 5%CO2It is incubated 0.5-2 hours in incubator. Collect cell suspension simultaneously centrifuge washing 2 times;Leave and take quality-control sample;Gained CTL is resuspended with physiological saline 100mL, and 0.5% people is added Blood albumin, standby human vein, which instils, to be used;Or it is resuspended using 30mL or 2mL according to clinical demand, standby torso model, intraperitoneal injection With or Partial tumors local injection.Trypan blu e dyes, and carries out cell count, 95% or more Cell viability;Total number of cells amount 1 ×1010Left and right;Flow cytometry cell phenotype shows that CD3+T cells account for 90% or more, and with CD8+ cell (cytotoxicities T cell) dominant, (Fig. 2A, Fig. 2 B).
The tumour that the DC vaccines of the present invention induce peripheral blood to obtain in vitro is related or specific CTL belongs to guarantor of the invention Protect range.
Not strong, Antigenic Target is incomplete, tumor patient swollen for existing tumour antigen immunogenicity for the DC vaccines of the present invention Tumor antigen be not easy to obtain etc. the problem of problem, DC and CTL amplification in vitro lazy weights in terms of tumour antigens preparation and DC and The closed question of the immune detection point activated in CTL Process of in vitro, it is proposed that total solution, it can be effectively in body Outer and Immune inducing in vivo specific for tumour antigen CTL, and can specific killing, no obvious toxic-side effects efficiently be generated to tumour.
Beneficial effects of the present invention embody in the following areas:
(1) in terms of preparing tumour antigen, after present invention arsenic trioxide targeted induction tumor stem cell apoptosis, with adding The method of heat handles tumour cell, both can also induce tumor cells expression heat shock protein with inducing apoptosis of tumour cell. Heat shock protein can reside in endocytoplasmic reticulum, can also be expressed in cell surface, be important tumour antigen molecule companion Companion can increase the immunogenicity of tumour antigen.
(2) in terms of the selection of tumour antigen, the available tumour antigen of DC vaccines of the present invention includes not only tumour cell Surface protein, epicyte protein, intracellular protein, further include in culture solution outside tumour cell after tumor cell culture amplification The vesica of row.Can full tumor-cell antigen be obtained from tumour cell different level to greatest extent in this way, Antigenic Target covers Lid is comprehensive, and it is anti-as antigen molecule companion and vesica to have taken into account soluble protein, lipopoteins, its heat shock protein being rich in Former graininess, increases the immunogenicity of the hybrid antigen;It simultaneously can be from autologous patient excision or biopsy tumor tissues, chest and abdomen Oedema oncocyte obtains antigen, has expanded the source of autologous tumor antigens, has met the requirement of individuation.
(3) in terms of the acquisition of peripheral blood collection capacity and DC cell concentrations, the present invention utilizes the low single core of initial amount peripheral blood Cell can harvest comparatively high amts and the DC compared with high maturity, have after culture amplification in 8-10 days and load tumour antigen Clinical manipulation is simple, and securely and reliably, on the one hand the features such as side reaction is small, high efficiency, it is big due to extracting to alleviate tumor patient Amount peripheral blood brings painful and to the adverse effect of body;On the other hand it reduces the production cost, ensure that treatment DC The sufficient dosage of cell.
(4) blocking DC cell surfaces PD-L1 to start t cell proliferation program to inhibit immunology, present invention joint Break DC immune tolerances using CD40 monoclonal antibodies and PD-L1 monoclonal antibodies, DC is activated with CD40 monoclonal antibodies, DC tables are closed with anti-PD-L1 monoclonal antibodies Face PD-L1.At present in terms of the immunosuppression mechanism research of PD-1/PDL-1 test points, what those skilled in the art were primarily upon It is expression programmed cell death molecule (PD-1) after T cell activation, tumour cell then expresses PD-L1 molecules (PD-1's matches in tissue Body), start the apoptosis of T cell if the two combines, this is one of important immunosuppression mechanism of tumor immune escape.So And i.e. high expression PD-L1 molecules after often having ignored DC loads tumour antigen and activating, when DC is contacted with T cell, completion tumour The apoptosis program of T cell may be started during antigen submission and T cell activation.The present invention is closed using anti-PD-L1 monoclonal antibodies The PD-L1 on the surfaces load tumour antigen DC, blocks this immunosuppression mechanism, immune anti-to activate body antineoplastic specificity It answers, is internal and external induced amplification tumor specific cytotoxicity T cell, and persistently play T cell effect and lay a good foundation.
(5) enhancing DC the vaccines reaction of Immune inducing in vivo tumor-specific immunity and external evoked amplification tumour-specific again In terms of CTL, the present invention using when load tumour antigen DC vaccines harvest, acquires same tumor patient for the second time in another way A small amount of peripheral blood detaches mononuclearcell.It is risen in the zero moment of non-adherent mononuclearcell culture, utilizes DC produced by the present invention Vaccine is mixed with the pure T cell in higher ratio and mononuclearcell group in vitro, and excluding, tumor patient is peculiar In the external superior microenvironment of immunosuppressive agent, antigen submission and activating T cell are completed.This method can be from 45ml peripheral bloods It originates mononuclearcell and obtains CTL total number of cells amount 1 × 1010Left and right, higher than the DC-CIK quantity (1 × 10 of usual method amplification9 Left and right);95% or more Cell viability;Cell surface can high expression CD3, CD8 molecule.These tumor-specific CTL adoptive transfers To patient's body can direct killing tumour cell can improve curative effect with DC vaccine use in conjunction.
Description of the drawings
Figure 1A-Figure 1B is that DC cell surfaces height of the present invention expresses CD80, CD83, CD86, HLA-DR typical case's collection of illustrative plates.
Fig. 2A-Fig. 2 B are that CTL cell surfaces height of the present invention expresses CD3, CD8 molecule typical case's collection of illustrative plates.
Specific implementation mode
Following embodiment is used for illustrating the present invention, but is not limited to the scope of the present invention.Unless otherwise specified, embodiment Used in the conventional means that are well known to those skilled in the art of technological means, biomaterial used is commercial goods.
The preparation of the DC vaccines of 1 efficient load tumour antigen of embodiment
1. tumor cell culture
Tumor tissues or tumor tissue biopsy sample of the tumour cell from patient itself operation excision, and have with patient There is the tumor cell line of same pathological diagnosis feature.Cell line can be the self-built system in laboratory, also be purchased from China national cell guarantor Tibetan center or U.S. ATCC carry out in vitro culture according to the cellular biology of tumor characteristic respectively.Usually using tire containing 10-15% The DMEM culture solutions of cow's serum, at 37 DEG C, 5%CO2It is cultivated in incubator, passage amplification.
2. induced apoptosis simultaneously expresses heat shock protein
When waiting for that cell growth to logarithmic phase, 80% is paved with bottom of bottle, drug-treated is carried out, arsenic trioxide (1- is added 15umol/L) continue at 37 DEG C, 5%CO2It is cultivated 4-24 hours in incubator.The diminution of microscopic observation tumour cell is rounded, and is pasted Wall rate reduces and cytoplasm " blebbing ", and cell is transferred to 43 DEG C of heating, 5%CO2It is cultivated 10 hours in incubator.
3. the preparation of tumour cell excretion body
After by arsenic trioxide and heating sequential induction, apoptosis occurs for tumour cell, and attached cell suspends, and outer row is more Excretion body.In order to harvest more antigens, can attached cell further be detached with cell scraping, then proceed to following procedure:
1) low-speed centrifugal 300-600g 10-15 minutes, tumour cell precipitation was collected to prepare tumor cell proteins class antigen (see step 4,5).
2) it collects supernatant high speed centrifugation 10000g 30 minutes, regathers the further ultracentrifugation 100000g of supernatant 60 and divide Clock collects precipitation, as tumour cell excretion body.
4. the preparation of tumor cell surface proteantigen, using surface protein extracts kit, Mem-PEP Plus Kit, Purchased from Thermo, Inc.USA.
5. the preparation of tumour cell Membrane protein antigen uses Membrane protein extraction kit, Pierce Cell Surface Protein lsolation Kit are purchased from Thermo, Inc.USA.It is operated according to commodity service manual, prepares tumor cell membrane Proteantigen.
6. the preparation of load tumour antigen DC vaccines
1) separation of peripheral blood mononuclear cells
Using it is known in the art that easy peripheral blood mononuclear cells separation method.The present embodiment is used with lower section Method:Aseptic collection peripheral blood 45ml, is gently added on the lymphocyte separation medium of proportion 1.077, is detached using Density gradient centrifugation Mononuclearcell.Centrifuge tube middle white cellular layer is drawn, is moved into the centrifuge tube containing physiological saline, cell is washed, abandons Clearly, serum-free DC cell culture mediums (BYN-PD701) are added, cell is resuspended, carry out cell count.
2) directional induction and amplification of DC
Cell is at 37 DEG C, 5%CO2Adhere-wall culture 2-6 hours in incubator, non-attached cell is sucked out, rh-IL-4, rh- is added GM-CSF 1000IU (final concentration), 37 DEG C, 5%CO2In incubator, directional induction and amplification DC cells 7-9 days.It is given birth to according to cell Long and amplification situation, individuation timely and appropriate discovery are added containing rh-IL-4, and the serum-free of rh-GM-CSF 1000IU (final concentration) is trained Support base (BYN-PD701).
3) the tumour antigen sensitization of DC and activation
DC was cultivated to 7-9 days, using the antigen from autologous patient tumour source, or according to the pathological examination knot of patient Matching corresponding mixed rumour antigen (the tumor cell surface proteantigen: tumour cell memebrane protein of 1/10 volume is added in fruit Antigen: tumour cell excretion body mass ratio 1: 1: 2), while Polyinosinic injection (final concentration 0.2-0.8 μ g/ml), anti-human is added CD40 monoclonal antibodies (whole 2-10 μ g/ml concentration), 37 DEG C, 5%CO2In incubator overnight.
4) closing of the surfaces DC PD-L1 molecules
DC is cultivated and sensitization was to 8-10 day, before harvesting DC, addition anti human PD-L 1 antibody (monarch grow directly from seeds object) 1- The PD-L1 molecules on the surface 10ug/ml (final concentration), closing DC, 37 DEG C, 5%CO2It is incubated 0.5-2 hours in incubator.
5) harvest of DC cells
After the closing of the surfaces DC PD-L1 molecules, culture solution (not abandoned containing cell) is sucked out, is blown and beaten and is rushed with ice physiological saline It washes and remains attached cell until falling off, collect simultaneously centrifuge washing 2 times of all cell suspensions;Cell physiological saline 1ml is resuspended standby Treatment is used.Leave and take quality-control sample, conventional detection microorganism, endotoxin simultaneously;Fluidic cell cell art detects DC cell surfaces The expression (Figure 1A -1B) of CD80, CD83, CD86, HLA-DR, by Figure 1A -1B visible DC cell surfaces height expression CD80, CD83、CD86、HLA-DR。
DC vaccines manufactured in the present embodiment detach the low starting in adherent mononuclearcell group from 45 milliliters of periphery anticoagulations DC precursors are measured, after culture amplification and antigen sensibilization, can finally harvest tumour antigen sensitization DC total quantitys about 1 × 108-2 ×108, the needs of dosage and an induced amplification tumor-specific CTL needed for 1-2 DC vaccine therapy can be met simultaneously.It should Method has clinical application easy to operate, securely and reliably, the features such as side reaction is small, high efficiency, is provided for clinic another excellent The DC vaccines of choosing and the new way prepared for tumor specific cell.
The preparation of the DC vaccines of 2 efficient load tumour antigen of embodiment
Tumour cell is detached from tumor patient chest, ascites, after culture amplification, disposably from tumour cell and tumour cell Excretion body in culture solution combines the preparation method for preparing tumour antigen.
1. collecting tumor patient hydrothorax or ascites 500-1000ml under aseptic condition, 300g is centrifuged 8 minutes, abandons supernatant.
2. physiological saline is added to be resuspended to 100ml, cell suspension is made.
3. pair weight density centrifugation:It is from the bottom to top:1st layer of 100% human lymphocyte separating liquid (density 1.077g/ ml);2nd layer of 75% lymphocyte separation medium (density 1.077g/ml);3rd confluent monolayer cells suspension.Centrifugation:700g, 20 minutes.
4. the cell for collecting the 1st interface and the 2nd interface, brine 2 times are sucked out respectively.
5. repetitive operation step 3, step 4 are primary.
6. the cell at the 2nd interface is tumour cell.
7. if tumour cell is enough, and desirable tumour cell is directly handled by embodiment 1, and autologous tumor is extracted obtain Membrane protein antigen.
8. can also be handled after the tumour cell in vitro culture of separation by embodiment 1, except extraction autologous tumor is thin Membrane-associated protein antigen, while extracting tumour cell vesica.
After prepared by tumour antigen, the method that can refer to embodiment 1 prepares tumour antigen sensitization DC vaccines and CTL.
3 tumour antigen sensitization of embodiment activates the external evoked amplification tumor-specific CTLs of DC
DC vaccines made from embodiment 1 and embodiment 2 are selected respectively.
1, on the day of DC vaccines harvest, second of 40~50 milliliters of heparin anti-coagulating of pumping, according to the 1 of 1 step 6 of embodiment) With operation 2), the not adherent mononuclearcell of fresh separated is sucked out, with 5: 1 ratio and DC vaccines obtained (with reference to implementation DC vaccines made from 2 method of example 1 or embodiment) mixing, it sets containing serum-free CTL primary culture mediums (BYN-PT331) culture bottle Interior overnight, anti-human CD3 monoclonal antibodies, anti-human CD28 monoclonal antibodies, rh-IL-2 is added in next day, at 37 DEG C, 5%CO2It is cultivated in incubator.Thereafter According to cell growth and amplification situation, individuation timely and appropriate discovery adds the serum-free amplification culture medium (BYN- containing rh-IL-2 PT332 CTL amplification cultivations) are carried out.
2, the harvest of CTL
CTL was cultivated to 11-14 days, and when cell log summit of growth, before CTL harvests, anti-human PD-1 Dan Ke are added Grand antibody 1-10ug/ml (final concentration), the PD-1 molecules on the surfaces closing CTL, 37 DEG C, 5%CO2It is incubated 0.5-2 hours in incubator. Collect cell suspension simultaneously centrifuge washing 2 times;Leave and take quality-control sample;CTL is resuspended with physiological saline 100ml, and it is white that 0.5% people's blood is added Albumen, standby human vein, which instils, to be used;Alternatively, being resuspended using 30ml or 2ml according to clinical demand, standby torso model or intraperitoneal injection With, or part is for locally injected into tumor.Trypan blu e dyes, and carries out cell count, 95% or more Cell viability;Total number of cells amount General 1 × 1010Left and right, high person is up to 2 × 1010;Tumor patient CTL cell surfaces can high expression CD3, CD8 molecule (Fig. 2A- 2B)。
Although above the present invention is described in detail with a general description of the specific embodiments, On the basis of the present invention, it can be modified or is improved, this will be apparent to those skilled in the art.Cause This, these modifications or improvements, belong to the scope of protection of present invention without departing from theon the basis of the spirit of the present invention.

Claims (10)

1. the preparation method of the DC vaccines of efficient load tumour antigen, which is characterized in that include the following steps:
(1) tumour antigen is prepared;
(2) it extracts anticoagulation from peripheral vein and detaches mononuclearcell, with adherent low initial amount cell directional induction and amplification DC cells;
(3) tumour antigen and activator sensitization is used to activate DC cells;
(4) DC vaccines are harvested after the PD-L1 molecules of closing DC cell surfaces.
2. preparation method according to claim 1, which is characterized in that the periphery of the tumour antigen and step (2) of step (1) Blood depends on the clinical feasibility and tumour antigen for obtaining autologous tumor tissue or cell from same individual or Different Individual Type;
Step (1) the tumour antigen type is selected from Tumor Tissue Lysates, tumor cell lysate, tumor cell surface albumen Antigen, tumour cell Membrane protein antigen, tumour cell excretion body, other tumours are related or specific proteins, tumour-specific are more It is one or more in peptide, the height personalization neoantigen based on gene sequencing.
3. preparation method according to claim 1, which is characterized in that step (1) described tumour antigen passes through chemical treatment It is obtained with the following methods of the sequential progress of physical heating:After arsenic trioxide culture is added into tumour cell, tumour cell is waited for Diminution is rounded, adherent rate reduces, after cytoplasm blebbing, 40-46 DEG C of culture cell, to induce the expression of heat shock protein.
4. preparation method according to claim 1, which is characterized in that prepare the DC vaccines per person-portion only needs in step (2) 40-50ml peripheral vein anticoagulations are extracted, with a large amount of DC of mononuclearcell induced amplification of low starting quantity.
5. according to any preparation methods of claim 1-4, which is characterized in that step (3) sensitization activates the side of DC cells Method is:Using the antigen in the autologous tumor source from peripheral blood donor, or according to the pathological examination of patient as a result, being added and disease The mixed rumour antigen for the corresponding tumour that reason testing result matches, while activator is added, it is incubated and obtains the DC of sensitization activation Cell;
The activator is bacterial product, complex immunity globulin, monoclonal antibody, micromolecular compound.
6. preparation method according to claim 5, which is characterized in that step (4) is that the DC cells after sensitization activation add Enter anti human PD-L 1 antibody, is incubated 0.5-2 hours at 30-40 DEG C, to close the PD-L1 molecules of DC cell surfaces.
7. the efficient load tumour antigen DC vaccines that any preparation methods of claim 1-6 are prepared, the DC epidemic diseases Seedling can be directly used for active immunity treatment, can induced amplification specific for tumour antigen CTL in vivo.
8. induced amplification tumour is related in vitro or in vivo or the purposes of specific CTL for DC vaccines described in claim 7.
9. purposes according to claim 8, which is characterized in that induction specific for tumour antigen CTL methods are in patient the Primary blood drawing prepares DC vaccines and harvests the same day of DC, and the separation of 40~50mL anticoagulation cirumferential bloods is extracted simultaneously second in same individual Not adherent mononuclearcell is sucked out with DC vaccines with 3-7 in adhere-wall culture:1 ratio mixing, is added CTL serum-free Primary cultures Base after exo-antigen is offered, is added anti-human CD3 monoclonal antibodies, anti-human CD28 monoclonal antibodies, rh-IL-2 and carries out CTL cultures;
It is changed to amplification culture medium after CTL starts cloning growth and carries out amplification cultivation;CTL is cultivated to cell log summit of growth When, before CTL harvests, the anti-human PD-1 monoclonal antibodies of final concentration of 1-10ug/ml, the PD-1 on the surfaces closing CTL is added Molecule is incubated 0.5-2 hours;Collect cell suspension and centrifuge washing;Leave and take quality-control sample;Gained CTL is resuspended with physiological saline, In case patient is transfused.
10. the DC vaccines described in claim 7 induce the tumour of peripheral blood acquisition related or specific CTL in vitro.
CN201810448476.XA 2018-05-11 2018-05-11 DC vaccine for efficiently loading tumor antigen and method for inducing and amplifying tumor antigen specific CTL (cytotoxic T lymphocyte) Active CN108379569B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201810448476.XA CN108379569B (en) 2018-05-11 2018-05-11 DC vaccine for efficiently loading tumor antigen and method for inducing and amplifying tumor antigen specific CTL (cytotoxic T lymphocyte)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201810448476.XA CN108379569B (en) 2018-05-11 2018-05-11 DC vaccine for efficiently loading tumor antigen and method for inducing and amplifying tumor antigen specific CTL (cytotoxic T lymphocyte)

Publications (2)

Publication Number Publication Date
CN108379569A true CN108379569A (en) 2018-08-10
CN108379569B CN108379569B (en) 2022-12-02

Family

ID=63071013

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810448476.XA Active CN108379569B (en) 2018-05-11 2018-05-11 DC vaccine for efficiently loading tumor antigen and method for inducing and amplifying tumor antigen specific CTL (cytotoxic T lymphocyte)

Country Status (1)

Country Link
CN (1) CN108379569B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111840528A (en) * 2020-06-17 2020-10-30 河北大学 Tumor vaccine of exosome combined immune checkpoint blocking agent and preparation method thereof
CN113403275A (en) * 2021-07-01 2021-09-17 北京百益宁医学科技有限责任公司 Preparation method of new antigen of specific target cancer species
CN113444687A (en) * 2021-05-31 2021-09-28 浙江圣希澳医学科技有限公司 DC vaccine and DC-CTL method for transfection of CD40L through cell-penetrating peptide mediated tumor antigen polypeptide sensitization
CN114149970A (en) * 2021-11-09 2022-03-08 因诺伟(北京)生物医疗科技有限公司 Preparation method and application of peripheral blood hematopoietic stem cell-derived sensitized dendritic cells
WO2024066080A1 (en) * 2022-09-30 2024-04-04 苏州系统医学研究所 Arsenic compound-based tumor vaccine, preparation method therefor, and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102091327A (en) * 2010-12-27 2011-06-15 蔡建辉 Preparation method of dendritic cell (DC) vaccine loaded with autologous tumor associated holoantigen
CN105695403A (en) * 2015-12-31 2016-06-22 深圳市中美康士生物科技有限公司 Killer immune cell culturing method and application thereof
CN106177931A (en) * 2016-08-25 2016-12-07 河北利同康生物科技有限公司 The double CTL high efficiency that blocks of immune detection point kills the preparation method of cell preparation

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102091327A (en) * 2010-12-27 2011-06-15 蔡建辉 Preparation method of dendritic cell (DC) vaccine loaded with autologous tumor associated holoantigen
CN105695403A (en) * 2015-12-31 2016-06-22 深圳市中美康士生物科技有限公司 Killer immune cell culturing method and application thereof
CN106177931A (en) * 2016-08-25 2016-12-07 河北利同康生物科技有限公司 The double CTL high efficiency that blocks of immune detection point kills the preparation method of cell preparation

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HU, ZILONG; MA, YUE; SHANG, ZHIYANG; 等.: "Improving immunotherapy for colorectal cancer using dendritic cells combined with anti-programmed death-ligand in vitro", 《ONCOLOGY LETTERS》 *
HU, ZILONG; MA, YUE; SHANG, ZHIYANG; 等.: "Improving immunotherapy for colorectal cancer using dendritic cells combined with anti-programmed death-ligand in vitro", 《ONCOLOGY LETTERS》, vol. 15, no. 4, 30 April 2018 (2018-04-30), pages 2 - 3 *
ZILONG HU等: "Improving immunotherapy for colorectal cancer using dendritic cells combined with anti-programmed death-ligand in vitro", 《ONCOLOGY LETTERS》, vol. 15, no. 4, 7 February 2018 (2018-02-07), pages 1 *
何晓顺,焦兴元: "《胆道肿瘤学前沿》", 30 September 2012, pages: 377 - 378 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111840528A (en) * 2020-06-17 2020-10-30 河北大学 Tumor vaccine of exosome combined immune checkpoint blocking agent and preparation method thereof
CN113444687A (en) * 2021-05-31 2021-09-28 浙江圣希澳医学科技有限公司 DC vaccine and DC-CTL method for transfection of CD40L through cell-penetrating peptide mediated tumor antigen polypeptide sensitization
CN113403275A (en) * 2021-07-01 2021-09-17 北京百益宁医学科技有限责任公司 Preparation method of new antigen of specific target cancer species
CN114149970A (en) * 2021-11-09 2022-03-08 因诺伟(北京)生物医疗科技有限公司 Preparation method and application of peripheral blood hematopoietic stem cell-derived sensitized dendritic cells
WO2024066080A1 (en) * 2022-09-30 2024-04-04 苏州系统医学研究所 Arsenic compound-based tumor vaccine, preparation method therefor, and application thereof

Also Published As

Publication number Publication date
CN108379569B (en) 2022-12-02

Similar Documents

Publication Publication Date Title
CN108379569A (en) The DC vaccines of efficient load tumour antigen and its method of induced amplification specific for tumour antigen CTL
CN102091327B (en) Preparation method of dendritic cell (DC) vaccine loaded with autologous tumor associated holoantigen
CN104711221B (en) Isolating immune cells and the method for extracting PRP are automated from adult peripheral blood
CN109575118A (en) It is used to prepare the polypeptide fragment and DC vaccine of DC vaccine
CN104845934B (en) Bleeding of the umbilicus CD34+Derived from hematopoietic precursor cells Dendritic Cells is prepared on a large scale method
CN104694473B (en) The method that immunocyte is extracted in automation from adult peripheral blood
CN105316287A (en) Method for long-term storage and resuscitation culture of adult peripheral blood mononuclear cell
CN103301449B (en) A kind of preparation method of large-scale culture dendritic cell vaccine and application thereof
CN106177931B (en) The double preparation methods for blocking CTL high efficiency killing cell preparation of immune detection point
CN104498434B (en) A kind of preparation method of a large amount of BMDCs, gained BMDC
CN104262459B (en) Acute monocytic leukemia-associated antigen MLAA-34 epitope polypeptide, and vaccine and pharmaceutical application of acute monocytic leukemia-associated antigen MLAA-34 epitope polypeptide
CN105861435A (en) In-vitro amplification method of natural killer cells (NK)
CN103405759B (en) Method for preparing tumor-specific DC vaccine by applying CD34+ cells of umbilical cord blood
CN104894072B (en) A kind of preparation method and applications of autologous natural killer cells propagation
CN106589133A (en) Preparation and applications of liver-cancer-specific CTL cells induced by new enhanced antigen combined polypeptide
CN105176926A (en) Method for amplifying NK cells through in-vitro cultivation
CN103173411A (en) Method and kit for preparing dendritic cells
CN105505871B (en) A kind of effective amplification CIK and improve the method that its specificity kills tumor ability
CN108642013A (en) From being detached in Cord blood after CD34 candidate stem cells expand culture, induction prepares Dendritic Cells method to one kind on a large scale
CN109153974A (en) Enhance the composition to abnormal cell lethality and its application
CN106795492A (en) A kind of preparation method of tumor-specific CTL
CN104288179B (en) Dendritic Cells and its preparation method and application
CN110358737A (en) A method of Chimeric antigen receptor T lymphocyte is prepared using excretion body
CN106565828B (en) Polypeptide for inducing DC-CIK cells and application thereof in tumor cell treatment
CN102510756A (en) Oncofetal antigen/immature laminin receptor peptides for the sensitization of dendritic cells for cancer therapy

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
GR01 Patent grant
GR01 Patent grant