WO2023164990A1 - Preparation method and use of autologous tumor-draining lymph node lymphocytes - Google Patents

Preparation method and use of autologous tumor-draining lymph node lymphocytes Download PDF

Info

Publication number
WO2023164990A1
WO2023164990A1 PCT/CN2022/085870 CN2022085870W WO2023164990A1 WO 2023164990 A1 WO2023164990 A1 WO 2023164990A1 CN 2022085870 W CN2022085870 W CN 2022085870W WO 2023164990 A1 WO2023164990 A1 WO 2023164990A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
lymph node
draining lymph
tumor
preparation
Prior art date
Application number
PCT/CN2022/085870
Other languages
French (fr)
Chinese (zh)
Inventor
姚燕丹
黄松音
王瑞
伍小华
杨彦嘉
鲍燕
Original Assignee
中山大学孙逸仙纪念医院深汕中心医院
中山大学孙逸仙纪念医院
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 中山大学孙逸仙纪念医院深汕中心医院, 中山大学孙逸仙纪念医院 filed Critical 中山大学孙逸仙纪念医院深汕中心医院
Publication of WO2023164990A1 publication Critical patent/WO2023164990A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2301Interleukin-1 (IL-1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2304Interleukin-4 (IL-4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2306Interleukin-6 (IL-6)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/24Interferons [IFN]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/25Tumour necrosing factors [TNF]
    • 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
    • C12N2501/515CD3, T-cell receptor complex
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/11Coculture with; Conditioned medium produced by blood or immune system cells
    • C12N2502/1121Dendritic cells
    • 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
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • 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
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • C12N2509/10Mechanical dissociation

Definitions

  • the invention relates to the technical field of cell culture, and more specifically, to a preparation method and application of autologous tumor-draining lymph node lymphocytes.
  • Solid tumors Unlike hematological malignancies with lineage markers, solid tumors are highly heterogeneous, making it difficult to find ideal targets for all tumor cells in solid tumors.
  • Adoptive cellular immunotherapy is one of the hot researches in the field of cancer treatment at present. Past studies have shown that it has the potential to be applied in both hematological malignancies and solid tumors to obtain significant curative effects. Among them, chimeric antigen receptor T, TIL and other therapies Diverse and remarkable advances have also been made in cancer therapy.
  • chimeric antigen receptor T therapy targeting a single tumor antigen often produces antigen loss or leads to recurrence of more malignant tumor clones, and this type of therapy has some shortcomings.
  • TIL therapy and chimeric antigen receptor T therapy belong to cellular immunotherapy, but TIL therapy has some significant advantages in the treatment of solid tumors.
  • Adoptive cell therapy (ACT) was first proposed by Professor Steve A. Rosenberg and his colleagues at the National Institutes of Health (NIH). For the first time, TIL was isolated from multiple mouse tumor models.
  • Adoptive cell therapy for systemic treatment of tumors The therapy collects infiltrating lymphocytes from the patient's tumor, cultures and expands them in vitro, and finally returns them to the patient.
  • TIL therapy has been demonstrated in many solid tumors, including breast cancer, ovarian cancer, melanoma renal cell carcinoma, non-small cell lung cancer, prostate cancer, bladder cancer, head and neck cancer, sarcoma, and pancreatic cancer.
  • TIL is composed of T cells with diverse T cell receptor (TCR) clones, which can recognize a series of tumor antigens, superior tumor The homing ability and low off-target toxicity endow it with unique advantages in the treatment of solid tumors.
  • TCR T cell receptor
  • TILs are obtained by shredding or enzymatically digesting tumors obtained from surgical resection of patients, culturing cytotoxic T cells from tumor tissues, and expanding the cells through continuous passage until they reach the order of 10 10 to 10 11 for reinfusion.
  • the antitumor activity of TILs in vivo is highly dependent on the expansion, persistence, and continued activity of adoptively transferred cells.
  • T cell products dominated by effector cells have stronger tumor cytotoxicity, these cells also tend to be terminal. differentiation and dysfunction.
  • some researchers have optimized the above culture protocol, using a combination of T cell activation 1, 2 and 3 signals (OKT3 antibody, agonistic CD137/4-1BB and IL-2) to culture TIL from tumor tissue has improved the success rate of culture. , but all TILs are obtained at the peripheral tissue level, which still cannot avoid the tendency of terminal differentiation and dysfunction after reinfusion, and the expansion, persistence and continuous activity of adoptively transferred cells still cannot be solved.
  • the prior art urgently needs a kind of adoptively transferred cells that can be used as a source of T cells in ACT to overcome the tendency of terminal differentiation and dysfunction after reinfusion in the prior art, so as to improve the expansion, persistence, and persistence of cells after adoptive transfer. active effect.
  • the present invention aims at overcoming at least one deficiency of the above-mentioned prior art, and provides a method for preparing autologous tumor-draining lymph node lymphocytes and its application, through which the autologous tumor-draining lymph node lymphocytes that can be used as adoptively transferred cells can be obtained , to improve the culture success rate of adoptively transferred cells, as well as the curative effect and persistence of effect after reinfusion.
  • An object of the present invention is to provide a method for preparing autologous tumor-draining lymph node lymphocytes, comprising the steps of:
  • A1 Obtain draining lymph node tissue from the patient, digest and separate it, and prepare draining lymph node mononuclear cells;
  • step A2 Culture the cells obtained in step A1, activate and expand the T cells contained therein, and obtain autologous tumor-draining lymph node lymphocytes.
  • the effect of adoptive T cell therapy is closely related to the degree of differentiation of T cells.
  • memory T cells with low degree of differentiation they last longer in the body and have a stronger anti-tumor effect.
  • CD8 + T cells exposed to different degrees of antigen stimulation directly affect cell survival, differentiation and therapeutic effect in vivo during cell expansion in vitro and after adoptive transfer therapy in vivo.
  • T memory stem cells T SCM
  • T CM central memory T cells
  • Lymph nodes are secondary lymphoid organs, and secondary lymphoid organs are the places where mature lymphocytes (T lymphocytes, B lymphocytes) settle. After the thymus matures, they circulate to the lymph nodes through blood circulation. In the lymph nodes of healthy people, T cells account for the majority of lymphocytes. 75%. Draining lymph nodes are the first lymph nodes that the primary tumor will inevitably pass through when lymph node metastasis occurs. T cells in lymph nodes can be divided into naive T cells (TN) that have not been exposed to antigens, and T memory stem cells (T SCMs ) that have been stimulated to varying degrees by antigens.
  • TN naive T cells
  • T SCMs T memory stem cells
  • T CM central memory T cells
  • T EM effector memory T cells
  • T EX exhausted T cells
  • PD-1 is both a marker of activation and exhaustion, but the co-expression of multiple inhibitory receptors is the main feature of exhaustion, so the exhausted T cell marker can be set to CD3 + PD-1 + Tim3 + LAG3 + .
  • T lymphocytes Under chronic infection or long-term tumor antigen stimulation, T lymphocytes will expand and differentiate into effector cells and memory cells that clear pathogens. Memory cells can survive for a long time to ensure that the body responds quickly to protect the body when it is attacked again by the same antigen, and TSCM cells have shown their strong potential for tumor treatment.
  • the inventors propose to obtain lymphocytes from draining lymph nodes, and As a source of T cells in ACT, it may be better than tumor tissue T cells. To this end, the inventors proposed a method for preparing autologous tumor-draining lymph node lymphocytes, and obtained corresponding autologous tumor-draining lymph node lymphocytes (LNL cells).
  • the preparation method is simple and easy to operate, and the prepared LNL cells are easy to culture, which can improve the success rate of cell culture, and as adoptively transferred cells (including TIL) can provide effective tumor killing effect after adoptive reinfusion, compared with other sources Adoptive transfer of cells has significant curative effect and long-lasting effect.
  • the preparation method can obtain T cells with a lower differentiation level and have been exposed to tumor antigens from lymph nodes, so as to increase the proportion of memory T cells in cell products, prolong the duration of in vivo treatment, and reduce T cell exhaustion.
  • the preparation method provided by the present application also exists as a culture method, which can improve the success rate of culture of adoptively transferred LNL cells, and overcome the defects in the prior art such as difficult culture of adoptively transferred cells.
  • the autologous tumor-draining lymph node lymphocytes are DC-induced autologous tumor-draining lymph node lymphocytes, further comprising the steps of:
  • DC cells from the peripheral blood of the same patient, pulse DC with tumor lysate, make DC present tumor antigen, and then co-culture with autologous tumor-draining lymph node lymphocytes in step A2 to obtain DC-induced autologous tumor-draining lymph node lymphocytes.
  • this application can also use IL-2 and tumor lysates presented by DCs to further stimulate the specific proliferation of lymphocytes, increase the amplification factor, and increase the The proportion of T cell subtypes that are conducive to persistence and curative effect after adoptive reinfusion.
  • step A1 specifically includes:
  • A11 Obtain draining lymph nodes by biopsy, remove excess fat, wash, shred, add to human lymphocyte serum-free medium containing collagenase I, collagenase III and DNase to incubate and digest;
  • step A2 specifically includes:
  • the initial medium is a mixture of human lymphocyte serum-free medium, recombinant human IFN- ⁇ and human AB serum or autologous plasma or serum substitute form;
  • the continuous expansion medium is human lymphocyte serum-free medium, recombinant Human IL-2 and human AB serum or autologous plasma or serum substitutes are mixed to form.
  • step A3 specifically includes:
  • peripheral blood of the same patient was extracted intravenously, centrifuged to remove the upper plasma, diluted with the same volume of normal saline, and then centrifuged with Ficoll density gradient of human lymphocyte separation medium to obtain peripheral blood mononuclear cells;
  • the DC medium is formed by mixing human lymphocyte serum-free medium and GM-CSF/IL-4;
  • A33 Use tumor lysate to pulse DC cells, and DCs are cultured and matured in the maturation medium and then co-cultured with autologous tumor-draining lymph node lymphocytes to obtain DC-induced autologous tumor-draining lymph node lymphocytes.
  • the DC maturation medium is human lymphocyte-free Serum medium, TNF ⁇ /IL-1 ⁇ /IL-6 are mixed to form.
  • Another object of the present invention is to provide the use of autologous tumor-draining lymph node lymphocytes obtained by the above preparation method in the preparation of drugs for preventing and treating tumors.
  • drugs for the prevention and treatment of tumors include adoptive cellular immunotherapy drugs.
  • Another object of the present invention is to provide the use of the autologous tumor-draining lymph node lymphocytes obtained by the above preparation method in the preparation of ACT therapy cells.
  • ACT therapy cells include TIL cells.
  • Another object of the present invention is to provide a cell preparation, which contains the above-mentioned autologous tumor-draining lymph node lymphocytes, and a pharmaceutically acceptable carrier or solvent.
  • the solvent is PBS solution.
  • Another object of the present invention is to provide adoptive cells for tumor therapy, which are derived from autologous tumor-draining lymph node lymphocytes.
  • adoptive reinfusion of LNL cells can achieve its tumor killing effect and inhibit tumor growth, and it has the function of adoptive infusion to treat tumors. Based on the source cells, it is expected to overcome the shortcomings of traditional TIL cells in the prior art.
  • the beneficial effect of the present invention is: through the preparation method provided by the application, autologous tumor-draining lymph node lymphocytes (LNL) can be obtained.
  • LNL lymph node lymphocytes
  • cells it is convenient to achieve in vitro culture expansion and improve the success rate of culture.
  • it can also significantly enhance the effect of in vivo expansion and the persistence and continuous activity of the in vivo effect, overcoming the use of tumor infiltration in the prior art. Defects such as terminal differentiation, dysfunction, and poor persistence that may result from T cells as a source.
  • the preparation method of the present application is simple to operate, has low requirements for experimental equipment, and is easy to be widely used in hospitals and other scientific research institutes. It is expected to become a new breakthrough in adoptive cellular immunotherapy programs including ACT therapy, and realize effective therapeutic preparations and treatment programs. So that more patients can be effectively treated.
  • Figure 1 shows the results of flow cytometry: showing the proportion and type of T lymphocytes from different tissues, including: T SCM : CD45RO - CD62L + CD95 + ; T CM : CD45RO + CD62L + ; T EM : CD45RO + CD62L - ; T EX : CD3+PD-1 + Tim3 + LAG3 + ; Th1: CD4 + T-bet + ; NK: CD3 - CD56 + .
  • Figure 2 shows the results of cell culture: showing the culture morphology and growth curve of LNL cells.
  • FIG. 3 shows the results of flow cytometry detection of cell subtypes on day 30 of LNL culture: T memory cells (T SCM , T CM , T EM ), Th1, NK and T lymphocyte exhaustion (T EX ).
  • Figure 4 shows the functional assessment of LNL at the cellular level: Tumor lysates or DCs stimulated with tumor lysates can stimulate the expression of CD134 and CD137, the markers of LNL activity in culture.
  • Figure 5 shows the curative effect of LNL on MDA-MB-231 tumor-bearing mice in Example 1: A, changes in body weight of mice during treatment; B, tumor growth curve; C, tumor suppression after treatment; D, tumor after treatment Infiltration of T cells in the tumor: IHC shows the infiltration of T cells in the tumor; E, the proportion of remaining T cells in the tumor cells after treatment.
  • Figure 6 shows the curative effect of LNL on PDX model mice in Example 1: A, changes in mouse body weight during treatment; B, tumor growth curve; C, tumor suppression after treatment (the red circle in the LNL treatment group represents complete regression of the tumor).
  • Figure 7 shows the main experimental process of Example 1.
  • test sample used in the following examples and the test process include the following (if the experimental specific conditions are not indicated in the examples, usually according to conventional conditions, or according to the conditions recommended by the reagent company; used in the following examples Reagents, consumables, etc., unless otherwise specified, can be obtained from commercial sources).
  • breast cancer is taken as an example to carry out the following experimental procedures.
  • the proportion and type of T lymphocytes in different tissue sources were detected by flow cytometry, as shown in Figure 1, the test results showed that T lymphocytes from different tissue sources There are significant differences in the proportion and type of cells. Compared with tumor-derived T cells, T cells in lymph nodes are more in a more immature memory cell state. On this basis, the applicant further carried out research on autologous tumor-draining lymph node lymphocytes and their use as a source of adoptively reinfused cells, hoping to overcome some of the shortcomings of other adoptively transferred cells in the prior art.
  • PBMC extract peripheral blood from a vein, centrifuge to remove the upper layer of plasma, and use the same method as step (5) to separate and prepare peripheral blood mononuclear cells with human lymphocyte separation medium; the acquisition of DC is to adhere to the wall for 2 hours after obtaining PBMC, and replace Fresh DC medium (Super Culture TM L500 human lymphocyte serum-free medium + 2U/ml GM-CSF/IL-4) was obtained from culture.
  • step 2 stimulate the cell suspension in step 2 with 100ng/mL CD3 monoclonal antibody and 1000IU/mL recombinant human IL-2 to activate and expand T cells; the T cells are stimulated by CD3 antibody After being activated, it is in the rapid expansion period.
  • DCs present tumor antigens to stimulate specific expansion of T cells
  • DCs loaded with tumor antigens were co-cultured with them to stimulate T cell-specific expansion.
  • the tumor tissue of the same patient was repeatedly frozen and thawed 5 times between liquid nitrogen and a 37°C water bath, centrifuged to remove cell debris, the lysate containing tumor antigens was placed in DC medium and pulsed for 24 hours, and DCs were stimulated by DC maturation factors. After 24 hours of maturation, DCs loaded with tumor antigens were co-cultured with T lymphocytes obtained after being stimulated and expanded by CD3 for 7 days;
  • the culture status of the isolated LNL cells in step 3 and step 4 is shown in Figure 2, where Day is the morphology on the second day of culture, REP-1W, REP-2W, and REP-3W are the culture values of the first week and the second day, respectively. Representative pictures of cell culture morphology at 1 week and 3 weeks.
  • the right of Figure 2 shows the corresponding growth curve of LNL cell culture, including CD3 monoclonal antibody and DC induction process.
  • LNL cells have been effectively cultured and expanded, and can continue to expand rapidly. That is, compared with other sources of adoptive cells in the prior art, LNL cells are easier to culture and expand, and can solve the problem that adoptive cells are difficult to cultivate and expand in large quantities in adoptive immune cell therapy in the prior art.
  • FIG. 3 shows the expression of exhaustion-related surface molecules (T EX ) of LNL T cells cultured in tumor-draining lymph nodes. After culturing until the 30th day, TEX accounted for only a very small proportion, less than 3%, indicating that the LNL cells provided in the examples have long-term efficacy and can effectively overcome the condition of being prone to terminal differentiation and exhaustion.
  • T EX exhaustion-related surface molecules
  • Cell preparation take the LNL cells (induced by DC) on the 14th day of culture, centrifuge at 750g for 5min, discard the supernatant, resuspend the cells with an appropriate volume of solvent (sterile PBS), count and adjust the cell concentration to 1 ⁇ 10 7 cells/125 ⁇ L, live cells shall prevail when counting cells.
  • solvent sterile PBS
  • the body weight of the mice in the treatment group was not significantly different from that of the IL-2 group and the PBS model group, indicating that LNL
  • the cell preparation has no obvious effect on the body weight of the experimental mice, and has certain safety.
  • the tumors of the mice in the LNL treatment group cultured in the tumor-draining lymph nodes were significantly inhibited and gradually subsided (as shown in Figures 5B and 5C), indicating that LNL cells have effective tumor-killing effects.
  • the results of immunohistochemistry showed the infiltration of lymphocytes in the tumor of the LNL group.
  • the LNL-derived cells were significantly infiltrated into the tumor;
  • the proportions of different subtypes of T cells in the tumor are shown in Figure 5E.
  • the CD3 + , CD4 + , and CD8 + subtype cells that almost disappeared in other groups the CD3 + , CD4 + , and CD8 + subtype cells in the LNL group all had an equivalent A large proportion is retained, indicating its long-term action characteristics.
  • the body weight of the mice in the treatment group had no statistical difference compared with the IL-2 group and the PBS model group, indicating that the LNL cell preparation had no significant effect on the body weight of the experimental mice. No significant impact, with a certain degree of security.
  • the tumors of the mice in the LNL treatment group cultured in the tumor-draining lymph nodes were significantly inhibited and gradually subsided (as shown in Figures 6B and 6C), indicating that LNL cells have specific and effective tumor-killing effects.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present invention provides a preparation method and the use of autologous tumor-draining lymph node lymphocytes. The preparation method comprises the following steps: A1, acquiring draining lymph node tissue from a patient, digesting same and separating same to prepare the draining lymph node mononuclear cells; and A2, culturing the cells obtained in the step A1, and activating and amplifying T cells contained in the cells to obtain the autologous tumor-draining lymph node lymphocytes. The preparation method of the present application can obtain the autologous tumor-draining lymph node lymphocytes (LNLs). Compared with tumor infiltrating lymphocytes, the LNLs as adoptive transfer cells facilitate in vitro culture amplification and increase the culture success rate. After adoptive transfer into a body, the LNLs can also provide a corresponding killing effect, and have excellent in-vivo amplification potential and in-vivo action durability and continuous activity, thus overcoming the defects of terminal differentiation, functional imbalance, poor durability and the like possibly caused by using tumor-infiltrating T cells as sources in the prior art.

Description

一种自体肿瘤引流淋巴结淋巴细胞的制备方法及其应用Preparation method and application of autologous tumor-draining lymph node lymphocytes 技术领域technical field
本发明涉及细胞培养技术领域,更具体地,涉及一种自体肿瘤引流淋巴结淋巴细胞的制备方法及其应用。The invention relates to the technical field of cell culture, and more specifically, to a preparation method and application of autologous tumor-draining lymph node lymphocytes.
背景技术Background technique
实体瘤与具有谱系标志物的血液系统恶性肿瘤不同,实体瘤具有高度异质性,因此,很难找到实体瘤中针对所有肿瘤细胞的理想靶点。过继性细胞免疫治疗是目前癌症治疗领域的热点研究之一,过往研究显示其在血液系统恶性肿瘤、实体瘤中均有应用以获得显著疗效的潜力,其中嵌合抗原受体T、TIL等疗法在肿瘤治疗方面也取得了多样、显著的进展。Solid tumors Unlike hematological malignancies with lineage markers, solid tumors are highly heterogeneous, making it difficult to find ideal targets for all tumor cells in solid tumors. Adoptive cellular immunotherapy is one of the hot researches in the field of cancer treatment at present. Past studies have shown that it has the potential to be applied in both hematological malignancies and solid tumors to obtain significant curative effects. Among them, chimeric antigen receptor T, TIL and other therapies Diverse and remarkable advances have also been made in cancer therapy.
对于实体瘤而言,嵌合抗原受体T疗法靶向单一肿瘤抗原通常会产生抗原丢失或导致更具恶性的肿瘤克隆复发,该类疗法还存在部分不足。TIL疗法与嵌合抗原受体T疗法同属于细胞免疫疗法,但TIL疗法在治疗实体瘤方面则具有一些显著的优势。过继细胞治疗(Adoptive cell therapy,ACT)最先由Steve A.Rosenberg教授和美国国立卫生院(NIH)的同事提出,首次从多个小鼠肿瘤模型中分离出TIL,是一种利用患者自身免疫系统治疗肿瘤的过继细胞疗法。该疗法通过从患者肿瘤中收集浸润的淋巴细胞,在体外进行培养和扩增,最后回输治疗患者。其作用原理是利用肿瘤浸润的淋巴细胞靶向多个肿瘤特异性抗原,且为MHC限制性,杀伤能力强,副作用小。TIL疗法的有效性已在许多实体瘤中得到验证,包括乳腺癌、卵巢癌、黑色素瘤肾细胞癌、非小细胞肺癌、前列腺癌、膀胱癌、头颈癌、肉瘤和胰腺癌。相较于其他过继细胞疗法(如嵌合抗原受体T和TCR-T疗法),TIL由具有多样化T细胞受体(TCR)克隆的T细胞组成,能够识别一系列肿瘤抗原,优越的肿瘤归巢能力和低脱靶毒性赋予其在治疗实体瘤方面的独特优势。For solid tumors, chimeric antigen receptor T therapy targeting a single tumor antigen often produces antigen loss or leads to recurrence of more malignant tumor clones, and this type of therapy has some shortcomings. TIL therapy and chimeric antigen receptor T therapy belong to cellular immunotherapy, but TIL therapy has some significant advantages in the treatment of solid tumors. Adoptive cell therapy (ACT) was first proposed by Professor Steve A. Rosenberg and his colleagues at the National Institutes of Health (NIH). For the first time, TIL was isolated from multiple mouse tumor models. Adoptive cell therapy for systemic treatment of tumors. The therapy collects infiltrating lymphocytes from the patient's tumor, cultures and expands them in vitro, and finally returns them to the patient. Its principle of action is to use tumor-infiltrating lymphocytes to target multiple tumor-specific antigens, and it is MHC-restricted, with strong killing ability and few side effects. The effectiveness of TIL therapy has been demonstrated in many solid tumors, including breast cancer, ovarian cancer, melanoma renal cell carcinoma, non-small cell lung cancer, prostate cancer, bladder cancer, head and neck cancer, sarcoma, and pancreatic cancer. Compared with other adoptive cell therapies (such as chimeric antigen receptor T and TCR-T therapy), TIL is composed of T cells with diverse T cell receptor (TCR) clones, which can recognize a series of tumor antigens, superior tumor The homing ability and low off-target toxicity endow it with unique advantages in the treatment of solid tumors.
但是,TIL的获得具有挑战性,体外培养自体肿瘤的成功率较低,且经体外长期培养及大量扩增后,T细胞体内持久性较差、会产生T细胞终末分化、功能障碍或衰竭等。目前TIL获取方法皆为从患者手术切除获得的肿瘤剪碎处理或经酶消化,从肿瘤组织中培养获得细胞毒性T细胞,并通过连续传代扩增细胞直至达到回输数量级10 10~10 11。但体内TIL的抗肿瘤 活性高度依赖于过继转移细胞的扩增、持久性和持续活性,虽然以效应型细胞为主导的T细胞产物具有更强的肿瘤细胞毒性,但这些细胞也倾向于终末分化和功能失调。虽有研究者优化了上述培养方案,用T细胞激活1、2和3信号(OKT3抗体、激动性CD137/4-1BB和IL-2)组合从肿瘤组织中培养TIL,提高了培养的成功率,但皆为外周组织水平获取TIL,仍然无法避免回输后终末分化、功能失调的倾向,过继转移细胞的扩增、持久性和持续活性问题仍无法解决。 However, the acquisition of TIL is challenging. The success rate of culturing autologous tumors in vitro is low, and after long-term culture and massive expansion in vitro, the persistence of T cells in vivo is poor, resulting in terminal differentiation, dysfunction or failure of T cells. wait. At present, TILs are obtained by shredding or enzymatically digesting tumors obtained from surgical resection of patients, culturing cytotoxic T cells from tumor tissues, and expanding the cells through continuous passage until they reach the order of 10 10 to 10 11 for reinfusion. However, the antitumor activity of TILs in vivo is highly dependent on the expansion, persistence, and continued activity of adoptively transferred cells. Although T cell products dominated by effector cells have stronger tumor cytotoxicity, these cells also tend to be terminal. differentiation and dysfunction. Although some researchers have optimized the above culture protocol, using a combination of T cell activation 1, 2 and 3 signals (OKT3 antibody, agonistic CD137/4-1BB and IL-2) to culture TIL from tumor tissue has improved the success rate of culture. , but all TILs are obtained at the peripheral tissue level, which still cannot avoid the tendency of terminal differentiation and dysfunction after reinfusion, and the expansion, persistence and continuous activity of adoptively transferred cells still cannot be solved.
因此,现有技术亟需一种可作为ACT中T细胞来源的过继转移细胞,以克服现有技术中回输后倾向终末分化、功能失调,以提高过继转移后扩增、持久性、持续活性效果。Therefore, the prior art urgently needs a kind of adoptively transferred cells that can be used as a source of T cells in ACT to overcome the tendency of terminal differentiation and dysfunction after reinfusion in the prior art, so as to improve the expansion, persistence, and persistence of cells after adoptive transfer. active effect.
发明内容Contents of the invention
本发明旨在克服上述现有技术的至少一种不足,提供一种自体肿瘤引流淋巴结淋巴细胞的制备方法及其应用,通过该制备方法能获得可应用为过继转移细胞的自体肿瘤引流淋巴结淋巴细胞,以提高过继转移细胞的培养成功率以及回输后的疗效和作用持久性。The present invention aims at overcoming at least one deficiency of the above-mentioned prior art, and provides a method for preparing autologous tumor-draining lymph node lymphocytes and its application, through which the autologous tumor-draining lymph node lymphocytes that can be used as adoptively transferred cells can be obtained , to improve the culture success rate of adoptively transferred cells, as well as the curative effect and persistence of effect after reinfusion.
本发明的一个目的在于提供一种自体肿瘤引流淋巴结淋巴细胞的制备方法,包括步骤:An object of the present invention is to provide a method for preparing autologous tumor-draining lymph node lymphocytes, comprising the steps of:
A1、从患者中获取引流淋巴结组织,消化、分离,制备引流淋巴结单个核细胞;A1. Obtain draining lymph node tissue from the patient, digest and separate it, and prepare draining lymph node mononuclear cells;
A2、对步骤A1获得的细胞进行培养,并激活、扩增其中包含的T细胞,获得自体肿瘤引流淋巴结淋巴细胞。A2. Culture the cells obtained in step A1, activate and expand the T cells contained therein, and obtain autologous tumor-draining lymph node lymphocytes.
过继的T细胞治疗效果与T细胞的分化程度息息相关,对于分化程度低的记忆T细胞,其体内存续时间长,有更强的抑瘤效果。而接触过抗原不同程度刺激的CD8 +T细胞在体外细胞扩增期间和体内过继转移治疗后直接影响体内细胞存续、分化和治疗效果。与效应记忆T细胞(T EM)和效应T细胞(T EFF)相比,T记忆干细胞(T SCM)和中枢记忆T细胞(T CM)具有更好的持久性和抗肿瘤免疫力,但如果经常受抗原和炎症信号刺激,T SCM将表现为多种抑制受体,如PD-1、TIM-3和LAG-3的表达,以及从氧化磷酸化到糖酵解的代谢变化,这些变化会导致T细胞耗竭。而具有和维持较少分化表型的T细胞的过继转移,对抗肿瘤疗效和患者预后至关重要。 The effect of adoptive T cell therapy is closely related to the degree of differentiation of T cells. For memory T cells with low degree of differentiation, they last longer in the body and have a stronger anti-tumor effect. However, CD8 + T cells exposed to different degrees of antigen stimulation directly affect cell survival, differentiation and therapeutic effect in vivo during cell expansion in vitro and after adoptive transfer therapy in vivo. Compared with effector memory T cells (T EM ) and effector T cells (T EFF ), T memory stem cells (T SCM ) and central memory T cells (T CM ) have better persistence and anti-tumor immunity, but if Frequently stimulated by antigenic and inflammatory signals, TSCM will manifest as expression of multiple inhibitory receptors, such as PD-1, TIM-3, and LAG-3, as well as metabolic changes from oxidative phosphorylation to glycolysis, which will lead to T cell exhaustion. The adoptive transfer of T cells with and maintaining a less differentiated phenotype is crucial for antitumor efficacy and patient prognosis.
淋巴结是次级淋巴器官,次级淋巴器官又是成熟淋巴细胞(T淋巴细胞、B淋巴细胞)定居的场所,在胸腺发育成熟后经血循环至淋巴结,健康人的淋巴结中T细胞占淋巴细胞的75%。引流淋巴结是原发肿瘤发生淋巴结转移必然会经过的第一批淋巴结,淋巴结中的T细胞可以分为未接触过抗原的初始T细胞(TN)、经抗原不同程度刺激的T记忆干细胞(T SCM)、中枢 记忆T细胞(T CM)、效应记忆T细胞(T EM)和处于耗竭的T细胞(T EX)。已知PD-1既是激活的标志又是耗竭的标志,但多种抑制性受体的共表达才是耗竭的主要特征,故可将耗竭的T细胞标志物设置为CD3 +PD-1 +Tim3 +LAG3 +。慢性感染或长期肿瘤抗原刺激下,T淋巴细胞会扩增并分化为清除病原体的效应细胞和记忆细胞。记忆细胞可以存活较长时间,以保障机体在受到相同抗原再次攻击时快速反应起保护作用,且T SCM细胞已显示出其强大的肿瘤治疗潜力。而肿瘤浸润的T细胞则大多数为趋于终末分化的效应T细胞,增殖能力弱,倾向耗竭,培养成功率低,在此基础上,本发明人提出从引流淋巴结中获取淋巴细胞,且作为ACT中T细胞来源可能更优于肿瘤组织T细胞。为此,本发明人提出了制备自体肿瘤引流淋巴结淋巴细胞的方法,并获得相应的自体肿瘤引流淋巴结淋巴细胞(LNL细胞)。该制备方法简单,易于操作,且制备获得的LNL细胞易于培养,能提高细胞培养成功率,作为过继转移细胞(包括TIL)在过继回输后能提供有效的肿瘤杀伤效果,相比于其他来源过继转移细胞,具有显著疗效和作用持久性。该制备方法能从淋巴器官淋巴结中获取较低分化水平且接触过肿瘤抗原的T细胞,以提高细胞制品中记忆T细胞的比例,延长体内治疗的存续时间,减少T细胞耗竭。且本申请提供的制备方法也作为一种培养方法而存在,能提高过继转移细胞LNL细胞的培养成功率,克服现有技术中过继转移细胞的培养难度大等缺陷。 Lymph nodes are secondary lymphoid organs, and secondary lymphoid organs are the places where mature lymphocytes (T lymphocytes, B lymphocytes) settle. After the thymus matures, they circulate to the lymph nodes through blood circulation. In the lymph nodes of healthy people, T cells account for the majority of lymphocytes. 75%. Draining lymph nodes are the first lymph nodes that the primary tumor will inevitably pass through when lymph node metastasis occurs. T cells in lymph nodes can be divided into naive T cells (TN) that have not been exposed to antigens, and T memory stem cells (T SCMs ) that have been stimulated to varying degrees by antigens. ), central memory T cells (T CM ), effector memory T cells (T EM ) and exhausted T cells (T EX ). It is known that PD-1 is both a marker of activation and exhaustion, but the co-expression of multiple inhibitory receptors is the main feature of exhaustion, so the exhausted T cell marker can be set to CD3 + PD-1 + Tim3 + LAG3 + . Under chronic infection or long-term tumor antigen stimulation, T lymphocytes will expand and differentiate into effector cells and memory cells that clear pathogens. Memory cells can survive for a long time to ensure that the body responds quickly to protect the body when it is attacked again by the same antigen, and TSCM cells have shown their strong potential for tumor treatment. However, most of the tumor-infiltrating T cells are effector T cells that tend to be terminally differentiated, have weak proliferative ability, tend to be exhausted, and have a low success rate of culture. On this basis, the inventors propose to obtain lymphocytes from draining lymph nodes, and As a source of T cells in ACT, it may be better than tumor tissue T cells. To this end, the inventors proposed a method for preparing autologous tumor-draining lymph node lymphocytes, and obtained corresponding autologous tumor-draining lymph node lymphocytes (LNL cells). The preparation method is simple and easy to operate, and the prepared LNL cells are easy to culture, which can improve the success rate of cell culture, and as adoptively transferred cells (including TIL) can provide effective tumor killing effect after adoptive reinfusion, compared with other sources Adoptive transfer of cells has significant curative effect and long-lasting effect. The preparation method can obtain T cells with a lower differentiation level and have been exposed to tumor antigens from lymph nodes, so as to increase the proportion of memory T cells in cell products, prolong the duration of in vivo treatment, and reduce T cell exhaustion. Moreover, the preparation method provided by the present application also exists as a culture method, which can improve the success rate of culture of adoptively transferred LNL cells, and overcome the defects in the prior art such as difficult culture of adoptively transferred cells.
进一步地,自体肿瘤引流淋巴结淋巴细胞为DC诱导的自体肿瘤引流淋巴结淋巴细胞,还包括步骤:Further, the autologous tumor-draining lymph node lymphocytes are DC-induced autologous tumor-draining lymph node lymphocytes, further comprising the steps of:
A3、从同一患者外周血获得DC细胞,并使用肿瘤裂解物脉冲DC,使DC呈递肿瘤抗原,然后与步骤A2中自体肿瘤引流淋巴结淋巴细胞共培养,获得DC诱导的自体肿瘤引流淋巴结淋巴细胞。A3. Obtain DC cells from the peripheral blood of the same patient, pulse DC with tumor lysate, make DC present tumor antigen, and then co-culture with autologous tumor-draining lymph node lymphocytes in step A2 to obtain DC-induced autologous tumor-draining lymph node lymphocytes.
本申请除了直接获得LNL细胞并利用其特性克服现有技术缺陷外,还能利用IL-2、DC递呈的肿瘤裂解物进一步刺激淋巴细胞特异性增殖,增大扩增倍数,并增加其中有利于过继回输后持久性、疗效的T细胞亚型比例。In addition to directly obtaining LNL cells and using their characteristics to overcome the defects of the prior art, this application can also use IL-2 and tumor lysates presented by DCs to further stimulate the specific proliferation of lymphocytes, increase the amplification factor, and increase the The proportion of T cell subtypes that are conducive to persistence and curative effect after adoptive reinfusion.
进一步地,步骤A1具体包括:Further, step A1 specifically includes:
A11、活检获取引流淋巴结,去除多余脂肪后洗涤,剪碎,再加入至含胶原酶I、胶原酶III和DNA酶的人淋巴细胞无血清培养基中孵育、消化;A11. Obtain draining lymph nodes by biopsy, remove excess fat, wash, shred, add to human lymphocyte serum-free medium containing collagenase I, collagenase III and DNase to incubate and digest;
A12、组织消化后用生理盐水稀释,使用细胞过滤器过滤细胞收集得到单细胞悬液;A12. Dilute the tissue with normal saline after digestion, and use a cell strainer to collect the cells to obtain a single cell suspension;
A13、获得单细胞悬液后,用人淋巴细胞分离液Ficoll密度梯度离心,吸取离心后以单个核细胞为主的白膜层,分离制备单个核细胞悬液,洗涤,种于孔板,获得引流淋巴结单个核细 胞。A13. After obtaining the mononuclear suspension, use the human lymphocyte separation medium Ficoll density gradient centrifugation, absorb the buffy coat layer mainly composed of mononuclear cells after centrifugation, separate and prepare the mononuclear cell suspension, wash, plant in the orifice plate, and obtain drainage Lymph node mononuclear cells.
进一步地,步骤A2具体包括:Further, step A2 specifically includes:
A21、将步骤A1获得的引流淋巴结单个核细胞加至初始培养基中培养,所述初始培养基为人淋巴细胞无血清培养基、重组人IFN-γ及人AB血清或自体血浆或血清替代物混合形成;A21. Add the draining lymph node mononuclear cells obtained in step A1 to the initial medium for culture, the initial medium is a mixture of human lymphocyte serum-free medium, recombinant human IFN-γ and human AB serum or autologous plasma or serum substitute form;
A22、经初始培养后,使用CD3单抗、重组人IL-2刺激,使T细胞激活并扩增;A22. After initial culture, stimulate with CD3 monoclonal antibody and recombinant human IL-2 to activate and expand T cells;
A23、待T细胞完全被活化后,定时更换一半的新鲜持续扩增培养基维持T细胞的增殖,获得自体肿瘤引流淋巴结淋巴细胞,所述持续扩增培养基为人淋巴细胞无血清培养基、重组人IL-2及人AB血清或自体血浆或血清替代物混合形成。A23. After the T cells are fully activated, regularly replace half of the fresh continuous expansion medium to maintain the proliferation of T cells, and obtain autologous tumor-draining lymph node lymphocytes. The continuous expansion medium is human lymphocyte serum-free medium, recombinant Human IL-2 and human AB serum or autologous plasma or serum substitutes are mixed to form.
进一步地,步骤A3具体包括:Further, step A3 specifically includes:
A31、静脉抽取同一患者的外周血,离心去上层血浆,同等体积生理盐水稀释后,用人淋巴细胞分离液Ficoll密度梯度离心,获得外周血单个核细胞;A31. The peripheral blood of the same patient was extracted intravenously, centrifuged to remove the upper plasma, diluted with the same volume of normal saline, and then centrifuged with Ficoll density gradient of human lymphocyte separation medium to obtain peripheral blood mononuclear cells;
A32、获得外周血单个核细胞后,贴壁培养,再更换DC培养基培养以获得DC细胞,所述DC培养基为人淋巴细胞无血清培养基、GM-CSF/IL-4混合形成;A32. After the peripheral blood mononuclear cells are obtained, culture them on the wall, and then replace the DC medium for culture to obtain DC cells. The DC medium is formed by mixing human lymphocyte serum-free medium and GM-CSF/IL-4;
A33、使用肿瘤裂解物脉冲至DC细胞,DC经成熟培养基培养成熟后与自体肿瘤引流淋巴结淋巴细胞共培养,获得DC诱导的自体肿瘤引流淋巴结淋巴细胞,所述DC成熟培养基为人淋巴细胞无血清培养基、TNFα/IL-1β/IL-6混合形成。A33. Use tumor lysate to pulse DC cells, and DCs are cultured and matured in the maturation medium and then co-cultured with autologous tumor-draining lymph node lymphocytes to obtain DC-induced autologous tumor-draining lymph node lymphocytes. The DC maturation medium is human lymphocyte-free Serum medium, TNFα/IL-1β/IL-6 are mixed to form.
本发明的又一目的在于提供上述制备方法所得自体肿瘤引流淋巴结淋巴细胞在制备预防、治疗肿瘤药物中的用途。Another object of the present invention is to provide the use of autologous tumor-draining lymph node lymphocytes obtained by the above preparation method in the preparation of drugs for preventing and treating tumors.
进一步地,预防、治疗肿瘤药物包括过继性细胞免疫治疗药物。Further, drugs for the prevention and treatment of tumors include adoptive cellular immunotherapy drugs.
本发明的再一目的在于提供上述制备方法所得自体肿瘤引流淋巴结淋巴细胞在制备ACT疗法细胞中的用途。ACT疗法细胞包括TIL细胞。Another object of the present invention is to provide the use of the autologous tumor-draining lymph node lymphocytes obtained by the above preparation method in the preparation of ACT therapy cells. ACT therapy cells include TIL cells.
本发明的再一目的在于提供一种细胞制剂,含有上述自体肿瘤引流淋巴结淋巴细胞,及药学上可接受的载体或溶剂。Another object of the present invention is to provide a cell preparation, which contains the above-mentioned autologous tumor-draining lymph node lymphocytes, and a pharmaceutically acceptable carrier or solvent.
进一步地,溶剂为PBS溶液。Further, the solvent is PBS solution.
本发明的再一目的在于提供一种肿瘤治疗用过继性细胞,来源于自体肿瘤引流淋巴结淋巴细胞。在本发明的一个实施例中,通过过继回输LNL细胞,能够实现其杀伤肿瘤的效果,抑制肿瘤生长,其具有过继回输以治疗肿瘤的功能。基于该来源细胞,有望克服现有技术传统TIL细胞的不足。Another object of the present invention is to provide adoptive cells for tumor therapy, which are derived from autologous tumor-draining lymph node lymphocytes. In one embodiment of the present invention, adoptive reinfusion of LNL cells can achieve its tumor killing effect and inhibit tumor growth, and it has the function of adoptive infusion to treat tumors. Based on the source cells, it is expected to overcome the shortcomings of traditional TIL cells in the prior art.
与现有技术相比,本发明的有益效果为:通过本申请提供的制备方法,能够获得自体肿 瘤引流淋巴结淋巴细胞(LNL),LNL相较于肿瘤浸润淋巴细胞而言,其作为过继回输细胞而言,便于实现体外的培养扩增,提高培养成功率,当过继转移至体内时,也能显著增强体内扩增效果以及体内作用的持久性、持续活性,克服现有技术中采用肿瘤浸润T细胞作为来源而可能导致的终末分化、功能失调、持久性差等缺陷。本申请制备方法操作简单,实验器材等要求低,便于广泛应用在医院等科研院所中,有望成为包括ACT疗法等过继性细胞免疫治疗方案上新的突破,实现有效治疗制剂、治疗方案等,使更多的患者得以有效的治疗。Compared with the prior art, the beneficial effect of the present invention is: through the preparation method provided by the application, autologous tumor-draining lymph node lymphocytes (LNL) can be obtained. As far as cells are concerned, it is convenient to achieve in vitro culture expansion and improve the success rate of culture. When adoptively transferred into the body, it can also significantly enhance the effect of in vivo expansion and the persistence and continuous activity of the in vivo effect, overcoming the use of tumor infiltration in the prior art. Defects such as terminal differentiation, dysfunction, and poor persistence that may result from T cells as a source. The preparation method of the present application is simple to operate, has low requirements for experimental equipment, and is easy to be widely used in hospitals and other scientific research institutes. It is expected to become a new breakthrough in adoptive cellular immunotherapy programs including ACT therapy, and realize effective therapeutic preparations and treatment programs. So that more patients can be effectively treated.
附图说明Description of drawings
图1显示流式结果:展示不同组织来源的T淋巴细胞占比及分型,其中含:T SCM:CD45RO -CD62L +CD95 +;T CM:CD45RO +CD62L +;T EM:CD45RO +CD62L -;T EX:CD3+PD-1 +Tim3 +LAG3 +;Th1:CD4 +T-bet +;NK:CD3 -CD56 +Figure 1 shows the results of flow cytometry: showing the proportion and type of T lymphocytes from different tissues, including: T SCM : CD45RO - CD62L + CD95 + ; T CM : CD45RO + CD62L + ; T EM : CD45RO + CD62L - ; T EX : CD3+PD-1 + Tim3 + LAG3 + ; Th1: CD4 + T-bet + ; NK: CD3 - CD56 + .
图2显示细胞培养结果:展示LNL细胞培养形态及生长曲线。Figure 2 shows the results of cell culture: showing the culture morphology and growth curve of LNL cells.
图3显示流式细胞术检测LNL培养第30天的细胞亚型结果:T记忆细胞(T SCM,T CM,T EM),Th1,NK及T淋巴细胞耗竭情况(T EX)。 Figure 3 shows the results of flow cytometry detection of cell subtypes on day 30 of LNL culture: T memory cells (T SCM , T CM , T EM ), Th1, NK and T lymphocyte exhaustion (T EX ).
图4显示细胞水平LNL的功能评估:肿瘤裂解物或经肿瘤裂解物刺激的DC能够刺激培养的LNL活性标志物CD134,CD137表达上调。Figure 4 shows the functional assessment of LNL at the cellular level: Tumor lysates or DCs stimulated with tumor lysates can stimulate the expression of CD134 and CD137, the markers of LNL activity in culture.
图5显示实施例1中LNL对MDA-MB-231荷瘤鼠的疗效:A、治疗期间小鼠体重变化;B、肿瘤生长曲线;C、治疗结束后肿瘤抑制情况;D、治疗结束后肿瘤中T细胞浸润情况:IHC显示肿瘤中T细胞浸润情况;E、治疗结束后肿瘤细胞中存留T细胞占比。Figure 5 shows the curative effect of LNL on MDA-MB-231 tumor-bearing mice in Example 1: A, changes in body weight of mice during treatment; B, tumor growth curve; C, tumor suppression after treatment; D, tumor after treatment Infiltration of T cells in the tumor: IHC shows the infiltration of T cells in the tumor; E, the proportion of remaining T cells in the tumor cells after treatment.
图6显示实施例1中LNL对PDX模型鼠的疗效:A、治疗期间小鼠体重变化;B、肿瘤生长曲线;C、治疗结束后肿瘤抑制情况(LNL治疗组红色圆圈代表肿瘤完全消退)。Figure 6 shows the curative effect of LNL on PDX model mice in Example 1: A, changes in mouse body weight during treatment; B, tumor growth curve; C, tumor suppression after treatment (the red circle in the LNL treatment group represents complete regression of the tumor).
图7显示实施例1主要实验过程。Figure 7 shows the main experimental process of Example 1.
具体实施方式Detailed ways
应该指出,以下详细说明都是例示性的,旨在对本申请提供进一步的说明。除非另有指明,本文使用的所有技术和科学术语具有与本申请所属技术领域的普通技术人员通常理解的相同含义。It should be pointed out that the following detailed description is exemplary and intended to provide further explanation to the present application. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this application belongs.
需要注意的是,这里所使用的术语仅是为了描述具体实施方式,而非意图限制根据本申 请的示例性实施方式。如在这里所使用的,除非上下文另外明确指出,否则单数形式也意图包括复数形式,此外,还应当理解的是,当在本说明书中使用术语“包含”和/或“包括”时,其指明存在特征、步骤、操作、器件、组件和/或它们的组合。It should be noted that the terminology used here is only for describing specific embodiments, and is not intended to limit the exemplary embodiments according to the present application. As used herein, unless the context clearly dictates otherwise, the singular is intended to include the plural, and it should also be understood that when the terms "comprising" and/or "comprising" are used in this specification, they mean There are features, steps, operations, means, components and/or combinations thereof.
现结合具体实例对本发明作进一步的说明,以下实施例仅是为了解释本发明,但不构成对本发明的限制。在以下实施例中所用到的试验样本及试验过程包括以下内容(如果实施例中未注明的实验具体条件,通常按照常规条件,或按照试剂公司所推荐的条件;下述实施例中所用的试剂、耗材等,如无特殊说明,均可从商业途径得到)。The present invention will now be further described in conjunction with specific examples. The following examples are only to explain the present invention, but not to limit the present invention. The test sample used in the following examples and the test process include the following (if the experimental specific conditions are not indicated in the examples, usually according to conventional conditions, or according to the conditions recommended by the reagent company; used in the following examples Reagents, consumables, etc., unless otherwise specified, can be obtained from commercial sources).
实施例1Example 1
在本实施例中,以乳腺癌为例进行以下实验过程。In this embodiment, breast cancer is taken as an example to carry out the following experimental procedures.
本实施例通过流式细胞术检测不同组织来源(相同体积的肿瘤和有肿瘤转移的引流淋巴结组织)中T淋巴细胞占比及分型,如图1所示,检测结果显示,不同组织来源T细胞的占比和分型具有显著差异,相比较肿瘤来源的T细胞,淋巴结中T细胞更多的处于更幼稚化的记忆细胞状态。在此基础上,本申请人进一步展开了自体肿瘤引流淋巴结淋巴细胞及其作为过继回输细胞来源的研究,以望克服现有技术其他过继转移细胞存在的部分不足。In this example, the proportion and type of T lymphocytes in different tissue sources (tumor with the same volume and draining lymph node tissue with tumor metastasis) were detected by flow cytometry, as shown in Figure 1, the test results showed that T lymphocytes from different tissue sources There are significant differences in the proportion and type of cells. Compared with tumor-derived T cells, T cells in lymph nodes are more in a more immature memory cell state. On this basis, the applicant further carried out research on autologous tumor-draining lymph node lymphocytes and their use as a source of adoptively reinfused cells, hoping to overcome some of the shortcomings of other adoptively transferred cells in the prior art.
具体地,展开有以下实验过程:Specifically, the following experimental procedures are developed:
1、单个核细胞获取:1. Mononuclear cell acquisition:
(1)活检获取引流淋巴结2-3个;(1) Biopsy to obtain 2-3 draining lymph nodes;
(2)利用解剖器械去除多余的脂肪后置于生理盐水中,重复洗3次;(2) Use dissecting instruments to remove excess fat, place in normal saline, and repeat washing 3 times;
(3)然后用无菌组织剪刀将肿瘤组织或淋巴结剪至约1-2mm 3大小的颗粒,加入混合了1.5mg/ml胶原酶I、III和0.2mg/ml DNA酶的Super Culture TML500人淋巴细胞无血清培养基中,37℃孵育半小时,每10min取出进行摇晃,消化组织块;进一步地,可采用全自动组织温和处理器实现该操作过程; (3) Then use sterile tissue scissors to cut the tumor tissue or lymph nodes to about 1-2mm 3 size particles, add Super Culture TM L500 mixed with 1.5mg/ml collagenase I, III and 0.2mg/ml DNase Lymphocyte serum-free medium, incubate at 37°C for half an hour, take out and shake every 10 minutes, and digest the tissue pieces; further, an automatic tissue gentle processor can be used to realize this operation process;
(4)组织消化后用生理盐水稀释,用无菌70μm细胞过滤器过滤细胞收集得到单细胞悬液,离心5min(2000rpm);(4) Dilute the tissue with normal saline after digestion, filter the cells with a sterile 70 μm cell filter to obtain a single cell suspension, and centrifuge for 5 min (2000 rpm);
(5)用人淋巴细胞分离液Ficoll密度梯度离心,水平离心20min(室温、800g、加减速度调为1),吸取中间以单个核细胞为主的白膜层分离制备单个核细胞悬液,生理盐水洗涤细胞2次后,计数,以2~2.5×10 6个/mL种于24孔板; (5) Use human lymphocyte separation medium Ficoll density gradient centrifugation, horizontal centrifugation for 20min (at room temperature, 800g, acceleration and deceleration speed adjusted to 1), absorb the buffy coat layer in the middle mainly mononuclear cells to separate and prepare mononuclear cell suspension, physiological After washing the cells twice with saline, count them, and seed them in a 24-well plate at 2-2.5× 106 cells/mL;
(6)PBMC:静脉抽取外周血,离心去上层血浆,采用步骤(5)相同的方法用人淋巴细胞分离液分离制备外周血单个核细胞;DC的获取是在获得PBMC后,贴壁2h,更换新鲜 DC培养基(Super Culture TML500人淋巴细胞无血清培养基+2U/ml GM-CSF/IL-4)培养获得。 (6) PBMC: extract peripheral blood from a vein, centrifuge to remove the upper layer of plasma, and use the same method as step (5) to separate and prepare peripheral blood mononuclear cells with human lymphocyte separation medium; the acquisition of DC is to adhere to the wall for 2 hours after obtaining PBMC, and replace Fresh DC medium (Super Culture TM L500 human lymphocyte serum-free medium + 2U/ml GM-CSF/IL-4) was obtained from culture.
2.T淋巴细胞初始培养2. Initial culture of T lymphocytes
在初始培养基:Super Culture TML500人淋巴细胞无血清培养基+10%人AB血清+1000U/mL的重组人IFN-γ中培养。 Cultured in the initial medium: Super Culture TM L500 human lymphocyte serum-free medium + 10% human AB serum + 1000 U/mL recombinant human IFN-γ.
3.T淋巴细胞激活和快速扩增3. T lymphocyte activation and rapid expansion
(1)培养7天后,用100ng/mL的CD3单抗和1000IU/mL的重组人IL-2刺激步骤2中的细胞悬液,使T细胞激活并扩增;所述T细胞经CD3抗体刺激后被激活处于快速扩增期。(1) After culturing for 7 days, stimulate the cell suspension in step 2 with 100ng/mL CD3 monoclonal antibody and 1000IU/mL recombinant human IL-2 to activate and expand T cells; the T cells are stimulated by CD3 antibody After being activated, it is in the rapid expansion period.
(2)每2~3天更换一半的新鲜培养基(新鲜培养基只添加重组人IL-2,未添加CD3单抗),并选择扩瓶,以保持细胞计数在2-2.5×10 6个/mL之间。 (2) Replace half of the fresh medium every 2 to 3 days (the fresh medium is only added with recombinant human IL-2, without adding CD3 monoclonal antibody), and choose to expand the bottle to keep the cell count at 2-2.5× 106 /mL.
4.DC呈递肿瘤抗原刺激T细胞特异性扩增4. DCs present tumor antigens to stimulate specific expansion of T cells
T细胞经CD3刺激扩增7天后,通过负载肿瘤抗原的DC与之共培养以刺激T细胞特异性扩增。具体的,将同一患者肿瘤组织在液氮和37℃水浴锅之间反复冻融5次,离心去除细胞碎片,将含有肿瘤抗原的裂解物置于DC培养基中脉冲24h,经DC成熟因子刺激DC成熟24h后,将负载肿瘤抗原的DC与前述经CD3刺激扩增7天后所得T淋巴细胞进行共培养;After T cells were stimulated and expanded by CD3 for 7 days, DCs loaded with tumor antigens were co-cultured with them to stimulate T cell-specific expansion. Specifically, the tumor tissue of the same patient was repeatedly frozen and thawed 5 times between liquid nitrogen and a 37°C water bath, centrifuged to remove cell debris, the lysate containing tumor antigens was placed in DC medium and pulsed for 24 hours, and DCs were stimulated by DC maturation factors. After 24 hours of maturation, DCs loaded with tumor antigens were co-cultured with T lymphocytes obtained after being stimulated and expanded by CD3 for 7 days;
每2~3天更换一半的新鲜的扩增培养基(Super Culture TML500人淋巴细胞无血清培养基+10%人AB血清+1000IU/mL的重组人IL-2)维持T细胞的增殖。 Half of the fresh expansion medium (Super Culture TM L500 human lymphocyte serum-free medium + 10% human AB serum + 1000 IU/mL recombinant human IL-2) was replaced every 2 to 3 days to maintain the proliferation of T cells.
步骤3及步骤4关于分离出的LNL细胞培养状态如图2所示,其中Day为培养第2天的形态,REP-1W、REP-2W、REP-3W分别为培养值第1周、第2周、第3周的细胞培养形态代表图。图2右显示了相应的LNL细胞培养生长曲线,包括CD3单抗、DC诱导过程,如图2右所示,LNL细胞得到有效的培养扩增,且能持续较快的扩增。即LNL细胞相比于现有技术中其他过继细胞来源,其更易于培养、扩增,能解决现有技术中过继免疫细胞疗法中过继细胞难以培养、大量扩增的不足。The culture status of the isolated LNL cells in step 3 and step 4 is shown in Figure 2, where Day is the morphology on the second day of culture, REP-1W, REP-2W, and REP-3W are the culture values of the first week and the second day, respectively. Representative pictures of cell culture morphology at 1 week and 3 weeks. The right of Figure 2 shows the corresponding growth curve of LNL cell culture, including CD3 monoclonal antibody and DC induction process. As shown in the right of Figure 2, LNL cells have been effectively cultured and expanded, and can continue to expand rapidly. That is, compared with other sources of adoptive cells in the prior art, LNL cells are easier to culture and expand, and can solve the problem that adoptive cells are difficult to cultivate and expand in large quantities in adoptive immune cell therapy in the prior art.
5.T细胞记忆亚群检测5. Detection of T cell memory subsets
(1)将LNL细胞连续培养至第30天取样,约10 5个/管,400g离心5min; (1) Continuously culture LNL cells until the 30th day and take samples, about 105 /tube, centrifuge at 400g for 5min;
(2)弃上清,加入500μL PBS,重悬细胞,400g离心5min;(2) Discard the supernatant, add 500μL PBS, resuspend the cells, and centrifuge at 400g for 5min;
(3)弃上清,加入100μL PBS,实验管分别标记相应荧光CD3,CD4,CD8,CD45RO,CD62L,CD95,CD56、T-bet抗体,室温避光孵育20min,设置阴性对照组;(3) Discard the supernatant, add 100 μL PBS, label the test tubes with corresponding fluorescent CD3, CD4, CD8, CD45RO, CD62L, CD95, CD56, T-bet antibodies, incubate at room temperature in the dark for 20 minutes, and set up a negative control group;
(4)加入500μL PBS,重悬细胞,400g离心5min,重复两次;(4) Add 500μL PBS, resuspend the cells, centrifuge at 400g for 5min, repeat twice;
(5)弃上清,加入100μL PBS,流式上机检测;(5) Discard the supernatant, add 100 μL PBS, and perform flow cytometry detection;
(6)用Flowjo软件分析,以CD3 +的细胞为分析对象设门,分析各亚群的比例(T SCM:CD45RO -CD62L +CD95 +;T CM:CD45RO +CD62L +;T EM:CD45RO +CD62L -(6) Analyze with Flowjo software, set a gate on CD3 + cells, and analyze the proportion of each subgroup (T SCM : CD45RO - CD62L + CD95 + ; T CM : CD45RO + CD62L + ; T EM : CD45RO + CD62L + - ;
(7)检测肿瘤引流淋巴结中培养的LNL记忆T细胞比例,检测结果如图3所示。CD3 +T细胞占细胞制剂的95%以上。具体地,中枢记忆T细胞、效应记忆T细胞占据较为显著的比例,表明了LNL细胞经培养至第30天,其仍然具有相应的分化、杀伤潜力。 (7) The proportion of LNL memory T cells cultured in the tumor draining lymph nodes was detected, and the detection results are shown in FIG. 3 . CD3 + T cells accounted for more than 95% of the cell preparations. Specifically, central memory T cells and effector memory T cells accounted for a relatively significant proportion, indicating that LNL cells still have corresponding differentiation and killing potential after cultured to day 30.
6.T细胞耗竭相关表面分子的检测6. Detection of surface molecules associated with T cell exhaustion
(1)培养至第30天取样;(1) Sampling after culturing until the 30th day;
(2)按上述T细胞记忆亚群检测过程收集方法收集样本,标记荧光抗体CD3、CD4、CD8、PD1、TIM3、LAG3,洗涤后100μL PBS重悬,流式上机检测;(2) Collect samples according to the collection method of T cell memory subpopulation detection process above, label fluorescent antibodies CD3, CD4, CD8, PD1, TIM3, LAG3, resuspend in 100 μL PBS after washing, and perform flow cytometry detection;
(3)用Flowjo软件分析,以CD3 +的细胞为分析对象设门,T细胞耗竭情况以PD1、TIM3、LAG3阳性比例表示; (3) Analyzed by Flowjo software, with CD3 + cells as the analysis object, and T cell exhaustion was expressed by the positive ratio of PD1, TIM3, and LAG3;
(4)如图3所示,显示了肿瘤引流淋巴结中培养的LNL T细胞耗竭相关表面分子表达情况(T EX)。培养至第30天,其中T EX仅占据非常小的比例,不足3%,表明实施例提供的LNL细胞,其具有长期作用效力,能有效克服容易终末分化、衰竭的状况。 (4) As shown in Figure 3, it shows the expression of exhaustion-related surface molecules (T EX ) of LNL T cells cultured in tumor-draining lymph nodes. After culturing until the 30th day, TEX accounted for only a very small proportion, less than 3%, indicating that the LNL cells provided in the examples have long-term efficacy and can effectively overcome the condition of being prone to terminal differentiation and exhaustion.
7.细胞水平LNL的功能评估7. Functional assessment of LNL at the cellular level
(1)LNL培养的第28天,按前述方法将同一患者肿瘤组织在液氮和37℃水浴锅之间反复冻融5次,离心去除细胞碎片,将含有肿瘤抗原的裂解物置于DC培养基中;(1) On the 28th day of LNL culture, the tumor tissue of the same patient was repeatedly frozen and thawed 5 times between liquid nitrogen and a 37°C water bath according to the above method, and the cell debris was removed by centrifugation, and the lysate containing tumor antigen was placed in DC medium middle;
(2)24h后,添加DC成熟因子培养24h;(2) After 24 hours, add DC maturation factors and culture for 24 hours;
(3)LNL细胞培养的第30天,更换新鲜的培养基(Super Culture TML500人淋巴细胞无血清培养基),以DC:LNL为1∶10~1∶100的比例置于DC培养瓶中,与成熟的DC共培养;或相同体积的肿瘤组织裂解物同一时间直接刺激同等计数的LNL;同时设置对照组:阴性对照组为相同计数的LNL更换新鲜培养基;阳性对照组为相同计数的LNL更换新鲜培养基并添加CD3抗体刺激; (3) On the 30th day of LNL cell culture, replace with fresh medium (Super Culture TM L500 human lymphocyte serum-free medium), and place it in DC culture flask at the ratio of DC:LNL 1:10~1:100 , co-cultured with mature DC; or the same volume of tumor tissue lysate directly stimulated the same count of LNL at the same time; set up the control group at the same time: the negative control group was replaced with fresh medium for the same count of LNL; the positive control group was the same count of LNL Replace LNL with fresh medium and add CD3 antibody stimulation;
(4)24h后观察DC形态及贴壁情况改变,按步骤5的方法收集样本,标记荧光抗体CD3,CD137,CD134;(4) After 24 hours, observe the changes in DC morphology and adherence, collect samples according to the method in step 5, and label fluorescent antibodies CD3, CD137, and CD134;
(5)洗涤后重悬,流式上机检测。结果如图4所示,肿瘤裂解物或经肿瘤裂解物刺激的DC能够有效刺激培养的LNL活性标志物CD134,CD137表达上调。(5) Resuspend after washing, and perform flow cytometry detection. The results are shown in Figure 4, tumor lysates or DCs stimulated by tumor lysates can effectively stimulate the cultured LNL activity marker CD134, and the expression of CD137 is up-regulated.
8.动物实验体内评估LNL细胞疗效及持久性8. In vivo evaluation of LNL cell efficacy and persistence in animal experiments
(1)细胞制剂:取培养第14天的LNL细胞(经DC诱导),750g离心5min后,弃上清,用适量体积的溶媒(无菌PBS)将细胞重悬,计数并调整细胞浓度为1×10 7个/125μL,细胞计数时以活细胞为准。 (1) Cell preparation: take the LNL cells (induced by DC) on the 14th day of culture, centrifuge at 750g for 5min, discard the supernatant, resuspend the cells with an appropriate volume of solvent (sterile PBS), count and adjust the cell concentration to 1×10 7 cells/125 μL, live cells shall prevail when counting cells.
(2)小鼠模型信息(2) Mouse model information
(a)种属&品系:NOD/SCID(a) Species & strains: NOD/SCID
动物等级:SPF级Animal grade: SPF grade
动物数量:15只Number of animals: 15
性别:雌Gender: female
年龄:4周龄Age: 4 weeks old
体重范围:17~22gWeight range: 17~22g
造模:MDA-MB-231细胞植入皮下成瘤Modeling: MDA-MB-231 cells implanted subcutaneously to form tumors
随机分组:PBS组、IL-2组、LNL组(每组5只)Random grouping: PBS group, IL-2 group, LNL group (5 rats in each group)
(b)种属&品系:NOD/SCID(b) Species & strains: NOD/SCID
动物等级:SPF级Animal grade: SPF grade
动物数量:12只Number of animals: 12
性别:雌Gender: female
年龄:4周龄Age: 4 weeks old
体重范围:17~22gWeight range: 17~22g
造模:原位肿瘤Modeling: Orthotopic Tumor
随机分组:PBS组、IL-2组、LNL组(每组4只)Random grouping: PBS group, IL-2 group, LNL group (4 rats in each group)
(3)治疗:细胞治疗组按1×10 7个/鼠尾静脉注射,体积为125μL,同时皮下辅以6000IU/125μL IL-2;其余对照组分别为尾静脉注射等体积的无菌PBS和皮下注射等体积的6000IU IL-2注射液。 (3) Treatment: In the cell therapy group, 1×10 7 cells/rat were injected into the tail vein with a volume of 125 μL, and at the same time, 6000 IU/125 μL IL-2 was subcutaneously supplemented; the other control groups were injected with the same volume of sterile PBS and An equal volume of 6000IU IL-2 injection was injected subcutaneously.
(4)安全及疗效(4) Safety and efficacy
a)监测小鼠体重变化、获得肿瘤生长曲线;a) Monitor the weight change of the mice and obtain the tumor growth curve;
b)监测治疗3-14周后小鼠肿瘤抑制情况;b) monitoring tumor suppression in mice after 3-14 weeks of treatment;
c)免疫组化检测PBS组、IL-2组、LNL组肿瘤淋巴细胞浸润情况(CD3)。c) Immunohistochemical detection of tumor lymphocyte infiltration (CD3) in PBS group, IL-2 group and LNL group.
d)流式细胞术检测治疗结束后PBS组、IL-2组、LNL组淋巴细胞比例;d) The proportion of lymphocytes in PBS group, IL-2 group and LNL group was detected by flow cytometry after treatment;
(5)安全及疗效结果(5) Safety and efficacy results
关于LNL对MDA-MB-231荷瘤鼠的安全和疗效(非特异),如图5A所示,治疗组小鼠的体重与IL-2组和PBS模型组比较均无统计学差异,表明LNL细胞制剂对实验鼠体重无明显影响,具有一定安全性。且肿瘤引流淋巴结中培养的LNL治疗组小鼠肿瘤明显抑制并逐渐消退(如图5B、5C所示),表明LNL细胞具有有效的肿瘤杀伤作用。治疗结束后,免疫组化结果显示LNL组肿瘤中淋巴细胞浸润情况,结果如图5D所示,LNL来源细胞得以显著浸润于肿瘤中;治疗结束后,流式细胞术检测LNL组治疗后小鼠肿瘤中T细胞不同亚型占比如图5E所示,相比于其他组近乎消失的CD3 +、CD4 +、CD8 +亚型细胞,LNL组CD3 +、CD4 +、CD8 +亚型细胞均有相当大比例留存,表明了其可长期作用的特性。 Regarding the safety and curative effect (non-specific) of LNL on MDA-MB-231 tumor-bearing mice, as shown in Figure 5A, the body weight of the mice in the treatment group was not significantly different from that of the IL-2 group and the PBS model group, indicating that LNL The cell preparation has no obvious effect on the body weight of the experimental mice, and has certain safety. Moreover, the tumors of the mice in the LNL treatment group cultured in the tumor-draining lymph nodes were significantly inhibited and gradually subsided (as shown in Figures 5B and 5C), indicating that LNL cells have effective tumor-killing effects. After the treatment, the results of immunohistochemistry showed the infiltration of lymphocytes in the tumor of the LNL group. As shown in Figure 5D, the LNL-derived cells were significantly infiltrated into the tumor; The proportions of different subtypes of T cells in the tumor are shown in Figure 5E. Compared with the CD3 + , CD4 + , and CD8 + subtype cells that almost disappeared in other groups, the CD3 + , CD4 + , and CD8 + subtype cells in the LNL group all had an equivalent A large proportion is retained, indicating its long-term action characteristics.
关于LNL对PDX模型鼠的安全和疗效(特异),如图6A所示,治疗组小鼠的体重与IL-2组和PBS模型组比较均无统计学差异,表明LNL细胞制剂对实验鼠体重无明显影响,具有一定安全性。且肿瘤引流淋巴结中培养的LNL治疗组小鼠肿瘤明显抑制并逐渐消退(如图6B、6C所示),表明LNL细胞具备特异性的且有效的肿瘤杀伤作用。Regarding the safety and efficacy (specificity) of LNL to PDX model mice, as shown in Figure 6A, the body weight of the mice in the treatment group had no statistical difference compared with the IL-2 group and the PBS model group, indicating that the LNL cell preparation had no significant effect on the body weight of the experimental mice. No significant impact, with a certain degree of security. Moreover, the tumors of the mice in the LNL treatment group cultured in the tumor-draining lymph nodes were significantly inhibited and gradually subsided (as shown in Figures 6B and 6C), indicating that LNL cells have specific and effective tumor-killing effects.
显然,本发明的上述实施例仅仅是为清楚地说明本发明技术方案所作的举例,而并非是对本发明的具体实施方式的限定。凡在本发明权利要求书的精神和原则之内所作的任何修改、等同替换和改进等,均应包含在本发明权利要求的保护范围之内。Apparently, the above-mentioned embodiments of the present invention are only examples for clearly illustrating the technical solution of the present invention, rather than limiting the specific implementation manner of the present invention. Any modification, equivalent replacement and improvement made within the spirit and principle of the claims of the present invention shall be included in the protection scope of the claims of the present invention.

Claims (10)

  1. 一种自体肿瘤引流淋巴结淋巴细胞的制备方法,其特征在于,包括步骤:A method for preparing autologous tumor-draining lymph node lymphocytes, comprising the steps of:
    A1、从患者中获取引流淋巴结组织,消化、分离,制备引流淋巴结单个核细胞;A1. Obtain draining lymph node tissue from the patient, digest and separate it, and prepare draining lymph node mononuclear cells;
    A2、对步骤A1获得的细胞进行培养,并激活、扩增其中包含的T细胞,获得自体肿瘤引流淋巴结淋巴细胞。A2. Culture the cells obtained in step A1, activate and expand the T cells contained therein, and obtain autologous tumor-draining lymph node lymphocytes.
  2. 根据权利要求1所述的制备方法,其特征在于,自体肿瘤引流淋巴结淋巴细胞为DC诱导的自体肿瘤引流淋巴结淋巴细胞,还包括步骤:The preparation method according to claim 1, wherein the autologous tumor-draining lymph node lymphocytes are DC-induced autologous tumor-draining lymph node lymphocytes, further comprising the steps of:
    A3、从同一患者外周血获得DC细胞,并使用肿瘤裂解物脉冲DC,使DC呈递肿瘤抗原,然后与步骤A2中自体肿瘤引流淋巴结淋巴细胞共培养,获得DC诱导的自体肿瘤引流淋巴结淋巴细胞。A3. Obtain DC cells from the peripheral blood of the same patient, pulse DC with tumor lysate, make DC present tumor antigen, and then co-culture with autologous tumor-draining lymph node lymphocytes in step A2 to obtain DC-induced autologous tumor-draining lymph node lymphocytes.
  3. 根据权利要求1所述的制备方法,其特征在于,步骤A1具体包括:The preparation method according to claim 1, wherein step A1 specifically comprises:
    A11、活检获取引流淋巴结,去除多余脂肪后洗涤,剪碎,再加入至含胶原酶I、胶原酶III和DNA酶的人淋巴细胞无血清培养基中孵育、消化;A11. Obtain draining lymph nodes by biopsy, remove excess fat, wash, shred, add to human lymphocyte serum-free medium containing collagenase I, collagenase III and DNase to incubate and digest;
    A12、组织消化后用生理盐水稀释,使用细胞过滤器过滤细胞收集得到单细胞悬液;A12. Dilute the tissue with normal saline after digestion, and use a cell strainer to collect the cells to obtain a single cell suspension;
    A13、获得单细胞悬液后,用人淋巴细胞分离液Ficoll密度梯度离心,吸取离心后以单个核细胞为主的白膜层,分离制备单个核细胞悬液,洗涤,种于孔板,获得引流淋巴结单个核细胞。A13. After obtaining the mononuclear suspension, use the human lymphocyte separation medium Ficoll density gradient centrifugation, absorb the buffy coat layer mainly composed of mononuclear cells after centrifugation, separate and prepare the mononuclear cell suspension, wash, plant in the orifice plate, and obtain drainage Lymph node mononuclear cells.
  4. 根据权利要求1所述的制备方法,其特征在于,步骤A2具体包括:The preparation method according to claim 1, wherein step A2 specifically comprises:
    A21、将步骤A1获得的引流淋巴结单个核细胞加至初始培养基中培养,所述初始培养基为人淋巴细胞无血清培养基、重组人IFN-γ及人AB血清或自体血浆或血清替代物混合形成;A21. Add the draining lymph node mononuclear cells obtained in step A1 to the initial medium for culture, the initial medium is a mixture of human lymphocyte serum-free medium, recombinant human IFN-γ and human AB serum or autologous plasma or serum substitute form;
    A22、经初始培养后,使用CD3单抗、重组人IL-2刺激;A22. After initial culture, stimulate with CD3 monoclonal antibody and recombinant human IL-2;
    A23、待T细胞完全被活化后,定时更换一半的新鲜持续扩增培养基维持T细胞的增殖,获得自体肿瘤引流淋巴结淋巴细胞,所述持续扩增培养基为人淋巴细胞无血清培养基、重组人IL-2及人AB血清或自体血浆或血清替代物混合形成。A23. After the T cells are fully activated, regularly replace half of the fresh continuous expansion medium to maintain the proliferation of T cells, and obtain autologous tumor-draining lymph node lymphocytes. The continuous expansion medium is human lymphocyte serum-free medium, recombinant Human IL-2 and human AB serum or autologous plasma or serum substitutes are mixed to form.
  5. 根据权利要求2所述的制备方法,其特征在于,步骤A3具体包括:The preparation method according to claim 2, wherein step A3 specifically comprises:
    A31、静脉抽取同一患者的外周血,离心去上层血浆,同等体积生理盐水稀释后,用人淋巴细胞分离液Ficoll密度梯度离心,获得外周血单个核细胞;A31. The peripheral blood of the same patient was extracted intravenously, centrifuged to remove the upper plasma, diluted with the same volume of normal saline, and then centrifuged with Ficoll density gradient of human lymphocyte separation medium to obtain peripheral blood mononuclear cells;
    A32、获得外周血单个核细胞后,贴壁培养,再更换DC培养基培养以获得DC细胞,所述DC培养基为人淋巴细胞无血清培养基、GM-CSF/IL-4混合形成;A32. After the peripheral blood mononuclear cells are obtained, culture them on the wall, and then replace the DC medium for culture to obtain DC cells. The DC medium is formed by mixing human lymphocyte serum-free medium and GM-CSF/IL-4;
    A33、使用肿瘤裂解物脉冲至DC细胞,DC经成熟培养基培养成熟后与自体肿瘤引流淋巴 结淋巴细胞共培养,获得DC诱导的自体肿瘤引流淋巴结淋巴细胞,所述DC成熟培养基为人淋巴细胞无血清培养基、TNFα/IL-1β/IL-6混合形成。A33. Use tumor lysate to pulse DC cells, and DCs are cultured and matured in the maturation medium and then co-cultured with autologous tumor-draining lymph node lymphocytes to obtain DC-induced autologous tumor-draining lymph node lymphocytes. The DC maturation medium is human lymphocyte-free Serum medium, TNFα/IL-1β/IL-6 are mixed to form.
  6. 权利要求1~5任一项所述制备方法所得自体肿瘤引流淋巴结淋巴细胞在制备预防、治疗肿瘤药物中的用途。The use of autologous tumor draining lymph node lymphocytes obtained by the preparation method described in any one of claims 1 to 5 in the preparation of drugs for preventing and treating tumors.
  7. 根据权利要求6所述的用途,其特征在于,预防、治疗肿瘤药物包括过继性细胞免疫治疗药物。The use according to claim 6, characterized in that the drugs for preventing and treating tumors include adoptive cellular immunotherapy drugs.
  8. 权利要求1~5任一项所述制备方法所得自体肿瘤引流淋巴结淋巴细胞在制备ACT疗法细胞中的用途。The use of autologous tumor draining lymph node lymphocytes obtained from the preparation method according to any one of claims 1 to 5 in the preparation of ACT therapy cells.
  9. 一种细胞制剂,其特征在于,含有权利要求1~5任一项所述制备方法获得的自体肿瘤引流淋巴结淋巴细胞,及药学上可接受的载体或溶剂。A cell preparation, characterized in that it contains autologous tumor-draining lymph node lymphocytes obtained by the preparation method according to any one of claims 1 to 5, and a pharmaceutically acceptable carrier or solvent.
  10. 一种肿瘤治疗用过继性细胞,其特征在于,来源于自体肿瘤引流淋巴结淋巴细胞。An adoptive cell for tumor treatment is characterized in that it is derived from autologous tumor draining lymph node lymphocytes.
PCT/CN2022/085870 2022-03-03 2022-04-08 Preparation method and use of autologous tumor-draining lymph node lymphocytes WO2023164990A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210204419.3A CN115433713B (en) 2022-03-03 2022-03-03 Preparation method and application of autologous tumor drainage lymph node lymphocyte
CN202210204419.3 2022-03-03

Publications (1)

Publication Number Publication Date
WO2023164990A1 true WO2023164990A1 (en) 2023-09-07

Family

ID=84240897

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/085870 WO2023164990A1 (en) 2022-03-03 2022-04-08 Preparation method and use of autologous tumor-draining lymph node lymphocytes

Country Status (2)

Country Link
CN (1) CN115433713B (en)
WO (1) WO2023164990A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101765607A (en) * 2007-05-31 2010-06-30 莱顿教学医院 HPV epitopes targeted by t cells infiltrating cervical malignancies for use in vaccines
CN104152411A (en) * 2014-08-08 2014-11-19 烟台博毓生物科技有限公司 Autologous dendritic cell activated tumor-infiltrating T-lymphocyte preparation method and application of T-lymphocyte
CN104946588A (en) * 2015-07-07 2015-09-30 英普乐孚生物技术(上海)有限公司 Separation and efficient amplification culture method for antigen specific T lymphocyte
US20170107490A1 (en) * 2014-06-11 2017-04-20 Polybiocept Ab Expansion of lymphocytes with a cytokine composition for active cellular immunotherapy
CN108220234A (en) * 2016-12-09 2018-06-29 贵州太瑞生诺生物医药有限公司 A kind of amplification in vitro method of the non-antitumor T cell in sentinel lymph node source
CN111801415A (en) * 2017-11-06 2020-10-20 路德维格癌症研究院 Method for expanding lymphocytes

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5814295A (en) * 1992-04-10 1998-09-29 The Ohio State University Research Foundation Determination of lymph nodes enriched in tumor reactive cells their proliferation and their use in adoptive cellular therapy
US20020177551A1 (en) * 2000-05-31 2002-11-28 Terman David S. Compositions and methods for treatment of neoplastic disease
US7651855B2 (en) * 2003-04-17 2010-01-26 The Trustees Of The University Of Pennsylvania Regulatory T cells and their use in immunotherapy and suppression of autoimmune responses
GB201504701D0 (en) * 2015-03-19 2015-05-06 Idogen Ab Novel treatment method
CN106795492A (en) * 2015-06-17 2017-05-31 深圳市达科为生物工程有限公司 A kind of preparation method of tumor-specific CTL
CN112831468A (en) * 2019-11-22 2021-05-25 路春光 TCM/TSCM cell for improving TIL cell induction and application
CN112972688A (en) * 2021-02-08 2021-06-18 吉林大学第一医院 Application of PPAR delta inhibitor and immunotherapy drug in preparation of antitumor drugs

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101765607A (en) * 2007-05-31 2010-06-30 莱顿教学医院 HPV epitopes targeted by t cells infiltrating cervical malignancies for use in vaccines
CN105031610A (en) * 2007-05-31 2015-11-11 莱顿教学医院 HPV epitopes targeted by t cells infiltrating cervical malignancies for use in vaccines
CN111686244A (en) * 2007-05-31 2020-09-22 莱顿教学医院 HPV epitopes targeted by T cells infiltrating cervical malignancies for use in vaccines
US20170107490A1 (en) * 2014-06-11 2017-04-20 Polybiocept Ab Expansion of lymphocytes with a cytokine composition for active cellular immunotherapy
CN104152411A (en) * 2014-08-08 2014-11-19 烟台博毓生物科技有限公司 Autologous dendritic cell activated tumor-infiltrating T-lymphocyte preparation method and application of T-lymphocyte
CN104946588A (en) * 2015-07-07 2015-09-30 英普乐孚生物技术(上海)有限公司 Separation and efficient amplification culture method for antigen specific T lymphocyte
CN108220234A (en) * 2016-12-09 2018-06-29 贵州太瑞生诺生物医药有限公司 A kind of amplification in vitro method of the non-antitumor T cell in sentinel lymph node source
CN111801415A (en) * 2017-11-06 2020-10-20 路德维格癌症研究院 Method for expanding lymphocytes

Also Published As

Publication number Publication date
CN115433713A (en) 2022-12-06
CN115433713B (en) 2023-10-27

Similar Documents

Publication Publication Date Title
JP6869994B2 (en) Medium addition kit for NK cell culture, and NK cell culture method using the kit
KR101909879B1 (en) Method and Composition for Proliferating of Natural Killer Cell
KR101503341B1 (en) Methods for isolation and proliferation of autologous cancer antigen-specific CD8+ T cells
KR101507174B1 (en) Immune privileged and modulatory progenitor cells
US20120107294A1 (en) Composition for in vivo transplantation for treatment of human cervical cancer comprising mononuclear cells derived from umblical cord blood
CN108697737A (en) The composition used in immunotherapy
JP6073417B2 (en) Spontaneous killing cell proliferation method and composition for spontaneous killing cell proliferation
Ferradini et al. Cytotoxic potential despite impaired activation pathways in T lymphocytes infiltrating nasopharyngeal carcinoma
CN108379569B (en) DC vaccine for efficiently loading tumor antigen and method for inducing and amplifying tumor antigen specific CTL (cytotoxic T lymphocyte)
CN110042080A (en) Separation, culture and the amplification in vitro method of primary carcinoma of liver tumor infiltrating lymphocyte
JP2021516666A (en) Prostate cancer-specific medullary infiltrative lymphocytes and their use
KR20140035102A (en) A METHOD FOR EXPANDING NK CELLS FROM PERIPHERAL BLOOD BELOW 100ml
CN103396992A (en) Culture and application of oligoclonal tumor-infiltrating lymphocytes for liver cancer
NZ569105A (en) A method for the expansion of tumor-reactive CD4 positive T-helper and CD positive T-lymphocytes
CN113663056A (en) Application of TNFSF15 protein as lymphocyte immunopotentiator and activation method thereof
US20210238549A1 (en) Use of memory lymphocyte population in liver cancer treatment
CN114209814A (en) Application of TNFSF15 protein in promoting differentiation of bone marrow stem cells into macrophages and amplification
CN117210411A (en) Immune cell and expression vector, application and preparation method thereof
WO2023164990A1 (en) Preparation method and use of autologous tumor-draining lymph node lymphocytes
WO2020198031A1 (en) Lung cancer specific marrow infiltrating lymphocytes and uses thereof
CN114989310A (en) Chimeric antigen receptor, macrophage for expressing chimeric antigen receptor and application
CN109666638A (en) Peripheral blood mononuclear cells is induced into the reagent and its composition therefor for CIK
CN113493525B (en) Synergistic and depletion resistant chimeric antigen receptor T cell and application thereof in preparation of tumor treatment drugs
CN114748457A (en) Pharmaceutical composition for treating cervical cancer and application thereof
CN110747167B (en) Preparation method and application of hemizygous BAK cell

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22929429

Country of ref document: EP

Kind code of ref document: A1