WO2008138214A1 - A method for activation and expansion of lymphocytes with high efficiency and the culture sysytem thereof - Google Patents
A method for activation and expansion of lymphocytes with high efficiency and the culture sysytem thereof Download PDFInfo
- Publication number
- WO2008138214A1 WO2008138214A1 PCT/CN2008/000887 CN2008000887W WO2008138214A1 WO 2008138214 A1 WO2008138214 A1 WO 2008138214A1 CN 2008000887 W CN2008000887 W CN 2008000887W WO 2008138214 A1 WO2008138214 A1 WO 2008138214A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- medium
- lymphocytes
- amplification
- cells
- concentration
- 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.)
- Ceased
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
- A61K40/428—Undefined tumor antigens, e.g. tumor lysate or antigens targeted by cells isolated from tumor
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/46—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0635—B lymphocytes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K2035/124—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the dose, timing or administration schedule
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/50—Cell markers; Cell surface determinants
- C12N2501/515—CD3, T-cell receptor complex
Definitions
- the present invention relates to the field of immunotherapy, and in particular to methods and culture systems for amplifying activated lymphocytes. Background technique
- LAK cells lymphokine activated killer cells
- the main effector cells of LAK cells are IL-2 activated NK cells. The earliest research found that these cells have the ability to kill leukemia cells in vitro.
- LAK cells belong to non-B non-T cells, this killing is non-HLA-restricted and represents the monitoring and killing effect of the body's natural immune system on tumor cells.
- NK cells have no killing effect on autologous leukemia cells without interferon (IFN), IL-2 activation, or removal of inhibitory monocytes.
- IFN interferon
- IL-2-activated T lymphocytes and NK cells secrete ⁇ -interferon, resulting in leukemia cells previously resistant to sputum cells being killed by activated sputum cells.
- LAK cells In addition to leukemia, some in vitro tests have shown that LAK cells also have a certain effect on animal and human malignant melanoma, kidney cancer, non-Hodgkin's lymphoma, lung cancer and colorectal cancer. However, due to the limitation of in vitro amplifiability and the effect of chemotherapy on lymphocytes, the anti-tumor effect of LAK cells in vivo is not satisfactory. In the early 1990s, Schmidt-Wolf IGH et al. (Schmidt-Wolf IG, Negrin RS, Kiem HP, Blume KG, Weissman IL.
- CIK cells differ from LAK cells in that the effector cells that exert anti-tumor effects are mainly CD3+CD56+ sputum cells (>50%). Studies have shown that CIK cells have significantly higher amplification efficiency, tumor cell killing ability, and activity in vivo than LAK cells.
- the traditional CIK treatment methods mainly have the following problems:
- the invention provides a method of amplifying activated lymphocytes, the method comprising:
- a lymphocyte activation step wherein the biological sample containing lymphocytes is contacted with a lymphocyte activator for a time and conditions sufficient to activate lymphocytes in the biological sample;
- a first amplification step wherein the activated lymphocytes obtained in the step (a) are cultured in a first medium containing a high concentration of IL-2;
- step (c) a second amplification step, wherein the lymphocytes obtained in the step (b) are cultured in a second medium containing a low concentration of IL-2;
- step (d) a third amplification step in which the lymphocytes obtained in the step (c) are cultured in a third medium containing a medium concentration of IL-2;
- the lymphocyte activation step and the first amplification step are performed simultaneously in the same culture vessel.
- the lymphocyte activator used in the method of the invention is an immobilized anti-CD3 antibody.
- the concentration of IL-2 in the first medium in step (b) may be, for example, 1000 U/ml to 6000 U/ml
- the second medium in step (c) The concentration of IL-2 may be, for example, 1 U/ml to 500 U/ml
- the concentration of IL-2 in the third medium in step (d) may be, for example, 300 U/ml to 750 U/ml.
- the medium of step (b) and / or (c) and / or (d) may also contain other cytokines, such as IL-7 and / or IL-15 o, preferably, the step (
- the concentration of IL-7 in the medium of b) and / or (c) and / or (d) may be, for example, 1-100 ng / ml, and the concentration of IL-15 may be, for example, 1-200 ng / ml.
- the culture media of steps (b), (c), and (d) may be the same or different.
- the first medium is RPMI-1640 medium
- the second medium is AIM-V medium
- the third medium is DMEM medium.
- the first, second, and third media may also be mixed media.
- the first amplification step described in the step (b) may preferably be carried out by bottle amplification, and the second expansion described in the step (c) may be carried out by bag amplification.
- the first amplification step described in step (b) is carried out for 7-14 days, and the second amplification step described in step (c) is carried out for 3 to 7 days, and in step (d)
- the third amplification step described is carried out for 3 to 7 days.
- step (d) may be repeated one or more times before step (e) is carried out.
- lymphocytes in the biological sample can be amplified 100 to 1000 times.
- the expanded cells are mainly CD8+ T cells and NK cells, for example, among the activated activated lymphocytes, CD8+ T cells can be >50%.
- the invention also provides a culture system for activating activated lymphocytes, comprising:
- an activation culture vessel for activating lymphocytes in a biological sample which comprises a lymphocyte activator
- a first amplification vessel for performing a first amplification step on activated lymphocytes comprising a first medium, said first medium comprising a high concentration of IL-2;
- a second amplification vessel for performing a second amplification step on the activated lymphocytes, comprising a second medium comprising a low concentration of IL-2;
- a third amplification vessel for performing a third amplification step on activated lymphocytes comprising a third medium containing a medium concentration of IL-2.
- the present invention also provides another culture system for amplifying activated lymphocytes, comprising:
- a first amplification container for activating a lymphocyte in a biological sample and simultaneously performing a first amplification step, comprising a lymphocyte activator and a first medium, said first medium containing a high concentration IL-2;
- a second amplification vessel for performing a second amplification step on the activated lymphocytes, comprising a second medium comprising a low concentration of IL-2;
- a third amplification vessel for performing a third amplification step on activated lymphocytes comprising a third medium containing a medium concentration of IL-2.
- the lymphocyte activator used in the culture system of the present invention is an immobilized anti-CD3 antibody.
- the concentration of IL-2 in the first medium may be, for example, 1000 U/ml to 6000 U/ml, and the concentration of IL-2 in the second medium. It may be, for example, 1 U/ml to 500 U/ml, and the concentration of IL-2 in the third medium may be, for example, 300 U/ml to 750 U/ml.
- the first and/or second and/or third medium may further contain other cytokines such as IL-7 and/or IL-15.
- cytokines such as IL-7 and/or IL-15.
- said first and / or The concentration of IL-7 in the di- and/or third medium may be, for example, 1-100 ng/ml, and the concentration of IL-15 may be, for example, 1-200 ng/ml.
- the first, second, and third media may be the same or different.
- the first medium is RPMI-1640 medium
- the second medium is AIM-V medium
- the third medium is DMEM medium.
- the first, second, and third media may also be a mixed medium.
- the present invention also provides an immunotherapy method comprising administering to an individual an expanded activated lymphocyte (EAL) obtained by the above method for amplifying activated lymphocytes of the present invention.
- EAL expanded activated lymphocyte
- the lymphocytes obtained by the above amplification method are Expanded Activated Autologous Lymphocytes (g ⁇ EAAL), and the immunotherapy method includes the following Steps:
- the immunotherapeutic methods of the invention are useful for treating individuals suffering from, for example, tumors, infectious diseases, congenital or acquired immunodeficiency.
- Tumors that can be treated using the immunotherapeutic methods of the invention are, for example, lung cancer, cardiac cancer, colon cancer, breast cancer, medulloblastoma, gastric cancer, renal cancer, and malignant melanoma, and individuals with tumors may be receiving Individuals who receive radiation and/or chemotherapy.
- Infectious diseases which can be treated using the immunotherapy method of the present invention are, for example, viral infections, particularly hepatitis B and C virus infections.
- Figure 1 Analysis of cell expansion folds before and after cell culture in 30 cases of 2-week activation/amplification culture.
- Figure 2 Analysis of lymphocyte phenotype before and after cell culture in 25 cases of 2-week activation/amplification culture.
- Figure 3 After EAAL reinfusion, the proportion of peripheral blood lymphocytes secreting IFN- ⁇ in patients is significantly increased.
- FIG. 6 Recurrent breast cancer, with decreased levels of tumor markers after treatment with EAAL of the invention (5 reinfusions). detailed description
- the present invention provides a method for efficiently amplifying activated lymphocytes and a culture system for the same, which is also called an Expanded Activated Lymphocyte Kit (EAL it) o
- the amplification method of the present invention is improved based on the conventional CIK method and is a highly efficient CIK cell culture method.
- the method of amplifying activated lymphocytes of the present invention comprises -
- lymphocyte activation step wherein the biological sample containing lymphocytes is contacted with a lymphocyte activator for a time and conditions sufficient to activate lymphocytes in the biological sample;
- step (b) a first amplification step in which the activated lymphocytes obtained in the step (a) are cultured in a first medium containing a high concentration of IL-2;
- step (c) a second amplification step, wherein the lymphocytes obtained in the step (b) are cultured in a second medium containing a low concentration of IL-2;
- step (d) a third amplification step in which the lymphocytes obtained in the step (c) are cultured in a third medium containing a medium concentration of IL-2;
- the lymphocyte activation step and the first amplification step are performed simultaneously in the same culture vessel.
- Lymphocytes that can be cultured and expanded using the methods of the invention can be derived from peripheral blood lymphocytes.
- the lymphocytes to be expanded may also be lymphocytes present in other biological samples, such as epithelial lymphocytes, intratumoral infiltrating lymphocytes, cancerous ascites or pleural immersion. Run lymphocytes and so on. Lymphocytes isolated from fresh peripheral blood are preferably used because of the ease of separation of lymphocytes in peripheral blood, high lymphocyte survival rate after isolation, and the like. Lymphocyte activation
- lymphocyte activation refers to the use of specific antigens or non-specific lymphocyte stimulators/molecules to stimulate lymphocytes, resulting in the synthesis of macromolecular substances (RNA, protein, DNA) and cytokines by lymphocytes. After lymphocyte activation, the cells proliferate and differentiate into progeny effector cells and memory cells of different functions.
- lymphocyte activator refers to a reagent/molecule that is capable of stimulating lymphocyte activation.
- an anti-CD3 antibody can be used as a lymphocyte activating agent which stimulates CD3 molecules on the surface of T cells and efficiently activates T lymphocytes through T cell receptors.
- Lymphocyte mitogens are another class of potent lymphocyte activators. These molecules rapidly activate lymphocytes and do not pass through T lymphocyte receptors.
- lymphocyte activator is an anti-CD3 antibody.
- the lymphocyte activating agent is an anti-CD3 antibody immobilized on a surface of a substrate.
- the use of an immobilized anti-CD3 antibody is preferred because it mainly activates CD8+ T cells having a cell killing function, which is particularly advantageous for enhancing the antitumor activity of the expanded lymphocytes.
- Methods of preparing culture vessels containing immobilized anti-CD3 antibodies are known and are described in detail below.
- the cytokines used to promote lymphocyte proliferation by the conventional lymphocyte expansion method are mainly IL-2, IL-4, IL-l and IFN- ⁇ .
- the compatible form of this cytokine primarily causes proliferation of CD3+CD56+ cells, CD4+ T cells, and CD8+ T cells.
- cytokines mainly use IL-2.
- IL-2 can amplify CD8+ T cells with high efficiency and specificity, and CD8+ T cells are the most important cellular components of the immune system to exert anti-tumor effects.
- the concentration of the cytokine IL-2 used in the conventional lymphocyte expansion method is always the same concentration from the start to the end of the culture.
- the present invention uses different concentrations of IL-2 in different culture stages.
- the activated lymphocytes obtained in the step (a) are cultured in the first medium containing the high concentration of IL-2 in the first amplification step (b).
- the concentration of IL-2 in the first medium ranges, for example, from 1000 U/ml to 6000 U/ml.
- the cells were grown to the 7-14th day in the first amplification step. Considering the nutrient consumption of the culture medium and the decrease in the activity of IL-2 and other cytokines, it is very important to add a proper amount of fresh culture solution every day.
- the lymphocytes obtained in the step (b) are further cultured in a second medium containing a low concentration of IL-2, so that the cells maintain the proliferation rate in the culture vessel. Not overactivated.
- the concentration of IL-2 in the second medium ranges, for example, from 1 U/ml to 500 U/ml.
- the cells are grown for 3-7 days in the second amplification step.
- the 10th-14th day of the culture enters a third amplification step (d) in which the lymphocytes obtained in the step (c) are cultured in a third medium containing a medium concentration of IL-2.
- the concentration of IL-2 in the third medium ranges, for example, from 300 U/ml to 750 U/ml.
- the cells are further cultured for 3 - 7 days in the third amplification step to extensively expand the cells to meet the clinical use requirements.
- lymphocytes can be amplified by a factor of 100 or more.
- IL-7 interleukin-7
- IL-15 interleukin-15
- the concentration of IL-7 in the medium of the step (b) and/or (c) and/or (d) is preferably from 1 to 100 ng/ml, and the concentration of IL-15 is preferably The ground is l-200ng/ml.
- the lymphocyte culture/amplification protocol used in the conventional method employs a fixed cell culture medium such as RPMI-1640 or DMEM.
- the medium of the steps (b), (c), and (d) may be different.
- the first medium is RPMI-1640 medium
- the second medium is AIM-V medium
- the third medium is DMEM medium.
- first, second, and third media may also be added Into other components that help lymphocyte expansion, such as fetal bovine serum or human serum.
- concentration of serum in the medium is 1 - 20%.
- the serum was thawed at 37 °C before use and inactivated by heating at 56 ° C for 30 minutes.
- mixed medium refers to a mixture of one or more of conventional media such as RPMI-1640, DMEM, IMEM, AIM-V, and the like. Training method
- any culture vessel suitable for lymphocyte activation and amplification can be used.
- the method of the present invention may employ a cell culture flask for cell culture, or a closed cell culture bag system for cell expansion and culture.
- the volume of the culture vessel should meet the need for extensive expansion of the cells.
- the culture bag which can be used in the present invention can, for example, accommodate the number of cells accommodated in 10-20 T225 cell culture flasks, which greatly reduces the workload of the cultured cells, while the closed system greatly reduces the possibility of contamination during the operation.
- the first amplification step can be performed by bottle amplification
- the second amplification step and the third amplification step can be performed by bag amplification.
- the lymphocyte activation step and the first amplification step are performed simultaneously in the same vial amplification vessel.
- the first amplification step is performed for 7-14 days
- the second amplification step is performed for 3 to 7 days
- the third amplification step is performed for 3 to 7 days.
- the third amplification step may be repeated one or more times before performing step (e).
- lymphocytes in the biological sample can be amplified at least 10 fold, preferably at least 100 fold, more preferably at least 1000 fold.
- the expanded cells are mainly CD8+ T cells and NK cells, for example, CD8+ T cells can be >50% in the expanded activated lymphocytes.
- the activation culture vessel is used to activate lymphocytes in a biological sample containing a lymphocyte activator.
- the activation culture vessel used in the culture/amplification system of the present invention contains an anti-CD3 antibody immobilized on an insoluble carrier such as the surface of the container as a lymphocyte activating agent.
- the anti-CD3 antibody can be diluted with a sterile sodium phosphate buffer (PBS) to a concentration of 1 - 50 w g/ml.
- PBS sterile sodium phosphate buffer
- the concentration is less than 1 ⁇ g/ml, the proliferation of T cells tends to be insufficient.
- the proliferation of T cells exceeds 50 ⁇ g/ml, the proliferation of T cells is not sufficient, but the free anti-CD3 antibody which is not immobilized on the insoluble carrier tends to increase.
- the diluent is not particularly limited as long as it is a physiological solution such as a sterilized sodium phosphate buffer solution, and as a protective agent, white protein, agarose or the like may be added.
- the diluted anti-CD3 antibody was aseptically plated on the bottom surface of the culture flask.
- the reaction temperature at the time of fixation is 4 to 40 ° C, more preferably 4 to 25 V, and most preferably 4 to 10 ° C. At less than 4 °C, the immobilization efficiency of the anti-CD3 antibody tends to decrease, and when it exceeds 40 °C, the activity of the anti-CD3 antibody tends to decrease.
- the reaction time in the immobilization is from 30 minutes to 24 hours, preferably from 1 hour to 24 hours. At less than 30 minutes, the immobilization amount of the anti-CD3 antibody tends to decrease, and when it exceeds 24 hours, the fixation time is too long, and the production efficiency tends to decrease.
- the residual anti-CD3 antibody dilution which was not immobilized was removed.
- the anti-CD3 antibody which has not been immobilized is washed with an appropriate washing solution such as sodium phosphate buffer (PBS) and dried.
- PBS sodium phosphate buffer
- the drying method can be carried out by natural drying in a 4-10 ° C cold storage, heating drying at 10-40 ° C, lyophilization, etc. after removing the cleaning liquid. In view of the activity and stability of the anti-CD3 antibody, lyophilization is preferred, and natural drying in a 4-10 °C cold storage is most preferred.
- the anti-CD3 antibody is immobilized on the surface of the insoluble carrier, and a carrier for T cell culture in which the immobilized surface of the anti-CD3 antibody is dried is obtained, and the culture vessel is obtained aseptically sealed.
- the first amplification vessel is used to perform a first amplification step on the activated lymphocytes, which contains a first medium containing a high concentration of IL-2.
- a concentration of IL-2 in the first medium is from 1000 U/ml to 6000 U/ml.
- the IL-2 concentration was diluted 1000-6000 U/ml with basal medium RPMI-1640 medium.
- IL-7 and/or IL-15 can be added.
- the concentrations were 1-100 ng/ml and 1-200 ng/ml, respectively.
- the medium is formulated into a cell culture medium having a volume ratio of 1 - 20% fetal calf serum or human serum (the serum is thawed at 37 ° C and heated at 56 ° C for 30 minutes), but In order to promote lymphocyte proliferation, it is generally preferably 2 to 10 (V/V)%.
- the culture medium is preferably a sterilized RPMI-1640 medium, but is not limited thereto, and other medium suitable for lymphocytes or a mixed medium can be used.
- the first medium for the first amplification vessel is thus prepared.
- the medium prepared as described above can be directly added to the above-described activated culture vessel.
- the second amplification vessel is for performing a second amplification step on the activated lymphocytes, which contains a second culture medium, and the second medium contains a low concentration of IL-2.
- the concentration of IL-2 in the second medium is from 1 U/ml to 500 U/ml.
- the second expansion vessel may use a bag culture vessel.
- the IL-2 concentration was diluted 1 - 500 U/ml with AIM-V medium.
- IL-7 and / or IL-15 o concentrations of 1-100 ng / ml and 1- 200 ng / ml can be added.
- the medium was formulated into a cell culture medium having a volume ratio of 1 - 20% fetal calf serum (the serum was thawed at 37 ° C and heated at 56 ° C for 30 minutes).
- a second medium for the second amplification vessel is thus prepared.
- the second culture medium preferably uses AIM-V medium, but is not limited thereto, and other medium or mixed medium suitable for lymphocytes may also be used.
- the third amplification vessel is used to perform a third amplification step on the activated lymphocytes, which contains a third medium containing a medium concentration of IL-2.
- a third medium containing a medium concentration of IL-2.
- the concentration of IL-2 in the third medium is from 300 U/ml to 750 U/ml.
- the third amplification container can use a bag culture container.
- the IL-2 concentration was diluted from DMEM medium to 300 - 750 U/ml.
- IL-7 and / or IL-15 can be added at concentrations of 1-100 ng/ml and 1- 200 ng/ml.
- the medium was formulated into a cell culture medium in a volume ratio of 1 - 20% fetal calf serum (the serum was thawed at 37 ° C and heated at 56 ° C for 30 minutes).
- the third medium for the third amplification vessel was thus prepared.
- the third medium is best used in DMEM medium, but it is not limited thereto, and other medium suitable for lymphocytes or mixed medium may also be used.
- the present invention also provides an immunotherapy method comprising administering to an individual an expanded activated lymphocyte (EAL) obtained by the method of amplifying activated lymphocytes of the present invention described above.
- EAL expanded activated lymphocyte
- the lymphocytes obtained by the above amplification method are Expanded Activated Autologous Lymphocytes (gp EAAL), and the immunotherapy method comprises the following steps :
- the amplified lymphocytes used are autologous lymphocytes, we call them Expanded Activated Autologous Lymphocytes (EAAL) Therapy.
- the lymphocytes used in the treatment are derived from a small amount of peripheral blood of the patient. These lymphocytes can be activated and amplified 100-1000 times in a short period of about 2 to 4 weeks and then returned to the patient.
- the immunotherapy method of the present invention can be used for treating an individual suffering from, for example, a tumor, an infectious disease, a congenital or acquired immunodeficiency defect, and an infectious disease caused by an organ transplant and an infection-induced tumor.
- the activated cells obtained by this method are mostly CD8+ T lymphocytes (>50%).
- CD8+ T lymphocytes play an important role in killing virus-infected cells and cancer cells.
- Human T lymphocytes include many specific cells directed against different antigens. These cells, after culturing, theoretically have broad recognition and killing effects against different tumor antigens/viral antigens. The purpose of treatment is to activate and proliferate such lymphocytes in vitro to enhance their function and to be used in therapy to kill tumor cells.
- Tumors that can be treated using the immunotherapeutic methods of the invention are, for example, lung cancer, cardiac cancer, colon cancer, breast cancer, medulloblastoma, gastric cancer, renal cancer, and malignant melanoma, and individuals with tumors may be receiving Individuals who receive radiation and/or chemotherapy.
- the immunotherapy method of the present invention can also be used to treat infectious diseases such as viral infections, particularly hepatitis B virus infection and hepatitis C virus infection.
- infectious diseases such as viral infections, particularly hepatitis B virus infection and hepatitis C virus infection.
- the culture system can be a standardized kit system
- the cells are stable, maintain cell viability within 24 hours after amplification, and can be returned within 12 hours;
- a culture flask coated with an immobilized anti-CD3 antibody was prepared.
- Anti-CD3 antibody (Ortho) was diluted to 10 wg/ml in sterile sodium phosphate buffer (PBS).
- PBS sterile sodium phosphate buffer
- the diluted anti-CD3 antibody was aseptically plated on the bottom of a culture flask (Costa). 4 ° C overnight. Wash the anti-CD3 antibody without immobilization with sodium phosphate buffer (PBS) and dry naturally in a cold storage at 4-10 °C. After drying, the immobilized surface of the anti-CD3 antibody was dried, and the carrier for T cell culture was aseptically sealed to obtain an activated culture vessel.
- the isolated lymphocytes were washed 3 times with RPMI-1640 medium (Invitrogen, Carlsbad, CA, USA), and finally a cell suspension having a cell density of about 2 ⁇ 10 6 /ml was prepared. 5 ml of this cell suspension (1 ⁇ 10 7 ) was added to the above activated culture vessel (culture flask coated with immobilized anti-CD3 antibody) in 50 ml of IL-2 containing 6000 U/ml as described above. The culture is started in the first amplification reagent.
- the cells and the medium were transferred to the second amplification reagent as described above after the 7th day of the culture, and growth was continued for 3 days in the cell culture bag.
- the cells in the second amplification reagent as described above were uniformly mixed with the third amplification reagent as described above, and then cultured in the two cell culture bags for 4 days. If a larger number of cells are required, the cells in the two cell culture bags can be further mixed with a reagent in the third amplification reagent as described above, and the culture is continued in three culture bags.
- the cells in the culture bag were transferred to a centrifuge tube, centrifuged at 1600 rpm for 10 minutes.
- the cells were washed twice with a solution of 1% albumin (Beijing Tiantan Biological Products Co., Ltd., China), and resuspended in 5% albumin-physiological saline for the following tests.
- the initial number of human lymphocytes cultured using the present invention was 20 ml of peripheral venous blood.
- the initial number of mononuclear cells before culture is about 0.5 - 5 X 10 7 .
- the number of cells is flat within two weeks. Both can be increased by 323 times (Fig. 1), and the cells can be amplified more than 1000 times after three weeks to four weeks of culture.
- the phenotypic detection method of lymphocytes is as follows:
- the labeled cells were 10 6 cells per tube, and the antibodies of the different combinations below were labeled according to the required cell phenotype.
- Cell markers were labeled with FITC-labeled anti-CD25, PE-CY5-labeled anti-CD4, FITC-labeled anti-CD16, PE-labeled anti-CD56, PE-CY5-labeled anti-CD8 (Becton-Dickinson) , Mountain View, CA, USA), PE-labeled anti-CD8 (ebioscience, San Diego, CA, USA) and ECD-labeled anti-CD3 (Beckman Coulter, Fullerton, CA, USA) o corresponding isotype antibody for Background correction. Phenotypic analysis was performed using a Coulter flow cytometer (COULTER EPICS XL) with the software COULTER EXP032 ADC (Beckman Coulter, Fullerton, CA, USA).
- the starting cells used in the present invention are peripheral blood mononuclear cells, wherein the T lymphocytes are about 40-70%, and the NK cells are about 1-5%.
- the main components of the cells are T lymphocytes and NK cells (>95%).
- the proportion of CD8+ T cells is about 50-95% (Fig. 2).
- CD8+ T lymphocytes and NK cells are the most important lymphocyte components of the body to exert anti-tumor and anti-viral effects.
- the ratio of CD4+ T cells and CD8+ T cells in the cells cultured by the method of the invention is significantly decreased, which is beneficial to the anti-tumor effect of lymphocytes.
- lymphocytes prepared according to the method of the present invention are returned to the patient, and the amount of the cells administered can be determined according to clinical needs, preferably 5 X 10 9 per cell.
- IFN- ⁇ intracellular cytokine Y interferon
- Lymphocytes were collected from patients receiving EAAL reinfusion. 2 X 10 6 /well cells were plated in 24-well plates using PMA (10 ng/ml; Sigma-Aldrich Corp., St. Louis, MO, USA) and Ionomycin (1 ⁇ g/ml; Sigma-Aldrich Corp. , St. Louis, MO, USA), stimulated for 6 hours in the presence of monensin (10 ⁇ M; Sigma-Aldrich Corp., St. Louis, MO, USA). Cells were harvested and stained with monoclonal antibodies against surface molecules such as ECD-CD3 and PE-CY5-CD8 for 15 minutes at room temperature.
- IFN- ⁇ In addition to its role in inhibiting and killing tumors, IFN- ⁇ also plays an important role in regulating the body's immune system. It can make the immune system's function tend to balance Thl, which is a killing function.
- Thl which is a killing function.
- the immune environment of the body is mostly an immunosuppressive environment, which hinders the killing of tumor cells by killer lymphocytes. Therefore, in addition to direct killing or inhibition of tumor growth, the mechanism of action of EAAL in anti-tumor therapy may include improving the immunosuppressive environment in patients and optimizing the immune environment for tumor killing (see (2) below).
- Immobilization, post-brain transfer, and radiotherapy Improvement in quality of life: After the cell was returned, it continued for one year of daily low heat loss, appetite and sleep improvement.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Immunology (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biochemistry (AREA)
- Hematology (AREA)
- General Engineering & Computer Science (AREA)
- Pharmacology & Pharmacy (AREA)
- Cell Biology (AREA)
- General Chemical & Material Sciences (AREA)
- Microbiology (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Description
高效扩增活化淋巴细胞的方法和培养系统 技术领域 Method and culture system for efficiently activating and activating lymphocytes
本发明涉及免疫治疗领域, 且具体涉及扩增活化淋巴细胞的方法和 培养系统。 背景技术 The present invention relates to the field of immunotherapy, and in particular to methods and culture systems for amplifying activated lymphocytes. Background technique
近年来, 由于免疫学的进步, 对由机体免疫监视能力的降低而产生 的各种免疫缺陷症己经进行了各种各样的免疫治疗方法的尝试。 由免疫 监视能力的降低而产生的各种免疫缺陷症包括癌症(恶性肿瘤)、 先天性 免疫缺陷、 后天性免疫缺陷等。 免疫疗法的目的之一是恢复或 /和增强免 疫监视能力, 从而对这些疾病进行治疗。 In recent years, due to advances in immunology, various immunotherapeutic methods have been tried for various immunodeficiency diseases caused by a decrease in the body's ability to monitor immunity. Various immunodeficiencies caused by a decrease in immune surveillance ability include cancer (malignant tumor), congenital immunodeficiency, acquired immunodeficiency, and the like. One of the purposes of immunotherapy is to restore or/and enhance the ability of immunosurveillance to treat these diseases.
作为癌症的免疫疗法, 细胞过继免疫治疗已成为控制肿瘤生长乃至 杀伤肿瘤细胞的一种重要的方法。 在二十世纪八十年代, 研究发现经 IL- 2体外刺激后, 外周血单个核细胞中的部分细胞可被特异性活化、 扩增, 并具有杀伤肿瘤细胞的作用。 这些细胞即淋巴因子活化的杀伤细胞(LAK 细胞) (Sinkovics JG, Horvath JC. Human natural killer cells: a comprehensive review. Int J Oncol. Jul 2005;27(l):5-47.)。 LAK细胞的主要效应细胞为 IL- 2活化的 NK细胞。 最早的研究发现, 这些细胞在体外具有杀伤白血病细 胞的能力。 由于 LAK细胞属于非 B非 T细胞, 因此, 这种杀伤是非 HLA 限制性的, 代表了机体天然免疫系统对肿瘤细胞的监视和杀伤作 用。 通常情况下, NK细胞不经干扰素(IFN)、 IL-2的活化、 或去除抑制 性单核细胞时, 对自体的白血病细胞没有杀伤作用。 经 IL-2活化的 T淋 巴细胞和 NK细胞可分泌 γ-干扰素, 导致以前对 ΝΚ细胞有抗性的白血病 细胞被活化的 ΝΚ细胞杀伤。 除白血病外, 一些体外试验表明, LAK细 胞对动物和人的恶性黑色素瘤、 肾癌、 非何杰金淋巴瘤、 肺癌及结直肠 癌也有一定的效果。 然而, 受到体外可扩增效率的限制、 化疗对淋巴细 胞的影响, LAK细胞在体内的抗肿瘤效果不理想。 九十年代初, Schmidt- Wolf IGH等人 ( Schmidt- Wolf IG, Negrin RS, Kiem HP, Blume KG, Weissman IL.Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med. 1991 Jul 1 ;174(1): 139-49. ) 发表了一种新的方法, 其在 体外培养外周血淋巴细胞, 然后过继回输用于治疗肿瘤病人。 与培养 LAK细胞不同, 这种方法使用了细胞因子 IL-2、 IL-4、 IL-1 α、 IFN- Υ、 抗 CD3抗体, 经过体外培养, 所获得的细胞对白血病和部分实体瘤有一 定的治疗效果。 为了与 LAK细胞相区别, 这种方法所获得的细胞被称为 细胞因子活化的杀伤细胞, 即 CIK细胞。 CIK细胞与 LAK细胞不同, 其 发挥抗肿瘤作用的效应细胞主要是 CD3+CD56+的 ΝΚΤ细胞(>50%)。 研 究表明, CIK细胞的扩增效率、 肿瘤细胞杀伤能力、 体内的活性比 LAK 细胞明显增高。 As an immunotherapy for cancer, cell adoptive immunotherapy has become an important method for controlling tumor growth and even killing tumor cells. In the 1980s, it was found that after stimulation with IL-2 in vitro, some cells in peripheral blood mononuclear cells can be specifically activated, expanded, and have the effect of killing tumor cells. These cells are lymphokine activated killer cells (LAK cells) (Sinkovics JG, Horvath JC. Human natural killer cells: a comprehensive review. Int J Oncol. Jul 2005; 27(l): 5-47.). The main effector cells of LAK cells are IL-2 activated NK cells. The earliest research found that these cells have the ability to kill leukemia cells in vitro. Since LAK cells belong to non-B non-T cells, this killing is non-HLA-restricted and represents the monitoring and killing effect of the body's natural immune system on tumor cells. Normally, NK cells have no killing effect on autologous leukemia cells without interferon (IFN), IL-2 activation, or removal of inhibitory monocytes. IL-2-activated T lymphocytes and NK cells secrete γ-interferon, resulting in leukemia cells previously resistant to sputum cells being killed by activated sputum cells. In addition to leukemia, some in vitro tests have shown that LAK cells also have a certain effect on animal and human malignant melanoma, kidney cancer, non-Hodgkin's lymphoma, lung cancer and colorectal cancer. However, due to the limitation of in vitro amplifiability and the effect of chemotherapy on lymphocytes, the anti-tumor effect of LAK cells in vivo is not satisfactory. In the early 1990s, Schmidt-Wolf IGH et al. (Schmidt-Wolf IG, Negrin RS, Kiem HP, Blume KG, Weissman IL. Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity J Exp Med. 1991 Jul 1 ;174(1): 139-49. ) A new method was developed for the culture of peripheral blood lymphocytes in vitro and then for subsequent treatment for cancer patients. Unlike LAK cells, this method uses cytokines IL-2, IL-4, IL-1 α, IFN-Υ, and anti-CD3 antibodies. After in vitro culture, the cells obtained have certain effects on leukemia and some solid tumors. The therapeutic effect. In order to distinguish from LAK cells, the cells obtained by this method are called cytokine-activated killer cells, that is, CIK cells. CIK cells differ from LAK cells in that the effector cells that exert anti-tumor effects are mainly CD3+CD56+ sputum cells (>50%). Studies have shown that CIK cells have significantly higher amplification efficiency, tumor cell killing ability, and activity in vivo than LAK cells.
传统的 CIK治疗方法主要存在以下问题: The traditional CIK treatment methods mainly have the following problems:
• 治疗前需要取大量病人外周血 (3000— 5000ml)以分离淋巴细胞, 对病人自身的免疫系统损伤较大; • Before treatment, a large number of patients' peripheral blood (3000-5000ml) should be taken to separate lymphocytes, which is harmful to the patient's own immune system;
• 培养效率低: 一般为 5— 10倍扩增; • Low efficiency of cultivation: generally 5-10 times amplification;
• 病人对放化疗耐受性差; • The patient is poorly tolerated by radiotherapy and chemotherapy;
• 细胞培养后, 含有较多的 CD4+CD25+的调节性 (抑制性) T细 胞, 回输后可能对体内的淋巴细胞增殖有一定的抑制性; • After cell culture, it contains more regulatory (inhibitory) T cells of CD4+CD25+, which may inhibit the proliferation of lymphocytes in vivo after reinfusion;
• 细胞稳定较差, 必须在扩增后 1一 2小时内使用; • The cells are poorly stable and must be used within 1 to 2 hours after amplification;
• 由于一次大量采集外周血单个核细胞, 该方法不能进行高频率治 疗; • Due to the large collection of peripheral blood mononuclear cells at one time, this method cannot be treated with high frequency;
• 病人需付出淋巴细胞分离的额外费用。 发明内容 • The patient is required to pay an additional fee for lymphocyte separation. Summary of the invention
在第一方面, 本发明提供一种扩增活化淋巴细胞的方法, 所述方法 包括: In a first aspect, the invention provides a method of amplifying activated lymphocytes, the method comprising:
(a)淋巴细胞活化步骤, 其中将含有淋巴细胞的生物学样品与淋巴细 胞活化剂相接触, 接触的时间和条件足以使得所述生物学样品中的淋巴 细胞发生活化; (b)第一个扩增步骤, 其中将步骤 (a)中得到的活化淋巴细胞在含有高 浓度 IL-2的第一培养基中进行培养; (a) a lymphocyte activation step, wherein the biological sample containing lymphocytes is contacted with a lymphocyte activator for a time and conditions sufficient to activate lymphocytes in the biological sample; (b) a first amplification step, wherein the activated lymphocytes obtained in the step (a) are cultured in a first medium containing a high concentration of IL-2;
(c)第二个扩增步骤, 其中将步骤 (b)中得到的淋巴细胞在含有低浓度 IL-2的第二培养基中进行培养; (c) a second amplification step, wherein the lymphocytes obtained in the step (b) are cultured in a second medium containing a low concentration of IL-2;
(d)第三个扩增步骤, 其中将步骤 (c)中得到的淋巴细胞在含有中等浓 度 IL-2的第三培养基中进行培养; 和 (d) a third amplification step in which the lymphocytes obtained in the step (c) are cultured in a third medium containing a medium concentration of IL-2;
(e)收集步骤 (d)中得到的扩增的活化淋巴细胞 (Expanded Activated Lymphocyte, 即 EAL)。 (e) Collecting Expanded Activated Lymphocytes (EAL) obtained in the step (d).
在本发明的方法的一个实施方式中, 淋巴细胞活化步骤与第一个扩 增步骤在同一个培养容器中同时进行。 In one embodiment of the method of the invention, the lymphocyte activation step and the first amplification step are performed simultaneously in the same culture vessel.
优选地, 本发明的方法中使用的淋巴细胞活化剂是固定化的抗 CD3 抗体。 Preferably, the lymphocyte activator used in the method of the invention is an immobilized anti-CD3 antibody.
优选地, 在本发明的方法中, 步骤 (b)中的第一培养基的 IL-2的浓度 可以是例如 1000 U/ml至 6000 U/ml, 步骤 (c)中的第二培养基的 IL-2的浓 度可以是例如 1 U/ml至 500 U/ml, 步骤 (d)中的第三培养基的 IL-2的浓度 可以是例如 300U/ml至 750 U/ml。 Preferably, in the method of the present invention, the concentration of IL-2 in the first medium in step (b) may be, for example, 1000 U/ml to 6000 U/ml, and the second medium in step (c) The concentration of IL-2 may be, for example, 1 U/ml to 500 U/ml, and the concentration of IL-2 in the third medium in step (d) may be, for example, 300 U/ml to 750 U/ml.
在本发明的方法中, 步骤 (b)和 /或 (c)和 /或 (d)的培养基中还可以含有 其他细胞因子, 例如 IL-7和 /或 IL-15 o 优选地, 步骤 (b)和 /或 (c)和 /或 (d) 的培养基中 IL-7的浓度可以是例如 l-100ng/ml, 而 IL-15的浓度可以是 例如 l-200ng/ml。 In the method of the invention, the medium of step (b) and / or (c) and / or (d) may also contain other cytokines, such as IL-7 and / or IL-15 o, preferably, the step ( The concentration of IL-7 in the medium of b) and / or (c) and / or (d) may be, for example, 1-100 ng / ml, and the concentration of IL-15 may be, for example, 1-200 ng / ml.
在本发明的方法中, 步骤 (b)、 (c)、 和 (d)的培养基可以是相同的或者 不同的。 优选地, 所述第一培养基是 RPMI-1640培养基, 所述第二培养 基是 AIM-V培养基, 所述第三培养基是 DMEM培养基。 所述第一、 第 二、 和第三培养基也可以是混和培养基。 In the method of the present invention, the culture media of steps (b), (c), and (d) may be the same or different. Preferably, the first medium is RPMI-1640 medium, the second medium is AIM-V medium, and the third medium is DMEM medium. The first, second, and third media may also be mixed media.
在本发明的方法中, 优选地可以通过瓶式扩增进行步骤 (b)中所述的 第一个扩增步骤, 并通过袋式扩增进行步骤 (c)中所述的第二个扩增步骤 和步骤 (d)中所述的第三个扩增步骤。 优选地, 步骤 (b)中所述的第一个扩 增步骤进行 7-14天, 步骤 (c)中所述的第二个扩增步骤进行 3至 7天, 且 步骤 (d)中所述的第三个扩增步骤进行 3至 7天。 在本发明的方法中, 在 进行步骤 (e)之前, 可以将步骤 (d)重复一或多次。 使用本发明的扩增活化淋巴细胞的方法, 所述生物学样品中的淋巴 细胞可被扩增 100至 1000倍。 优选地, 其中扩增后的细胞主要为 CD8+ T细胞和 NK细胞, 例如, 所扩增的活化淋巴细胞中, CD8+ T细胞可 >50%。 In the method of the present invention, the first amplification step described in the step (b) may preferably be carried out by bottle amplification, and the second expansion described in the step (c) may be carried out by bag amplification. The addition step and the third amplification step described in step (d). Preferably, the first amplification step described in step (b) is carried out for 7-14 days, and the second amplification step described in step (c) is carried out for 3 to 7 days, and in step (d) The third amplification step described is carried out for 3 to 7 days. In the method of the present invention, step (d) may be repeated one or more times before step (e) is carried out. Using the method of amplifying activated lymphocytes of the present invention, lymphocytes in the biological sample can be amplified 100 to 1000 times. Preferably, wherein the expanded cells are mainly CD8+ T cells and NK cells, for example, among the activated activated lymphocytes, CD8+ T cells can be >50%.
在第二方面, 本发明还提供一种用于扩增活化淋巴细胞的培养系 统, 其包括: In a second aspect, the invention also provides a culture system for activating activated lymphocytes, comprising:
(a)用于活化生物学样品中的淋巴细胞的活化培养容器, 其含有淋巴 细胞活化剂; (a) an activation culture vessel for activating lymphocytes in a biological sample, which comprises a lymphocyte activator;
(b)用于对活化淋巴细胞进行第一个扩增步骤的第一扩增容器, 其含 有第一培养基, 所述第一培养基含有高浓度的 IL-2; (b) a first amplification vessel for performing a first amplification step on activated lymphocytes, comprising a first medium, said first medium comprising a high concentration of IL-2;
(c)用于对活化淋巴细胞进行第二个扩增步骤的第二扩增容器, 其含 有第二培养基, 所述第二培养基含有低浓度的 IL-2; 和 (c) a second amplification vessel for performing a second amplification step on the activated lymphocytes, comprising a second medium comprising a low concentration of IL-2;
(d)用于对活化淋巴细胞进行第三个扩增步骤的第三扩增容器, 其含 有第三培养基, 所述第三培养基含有中等浓度的 IL-2。 (d) A third amplification vessel for performing a third amplification step on activated lymphocytes, comprising a third medium containing a medium concentration of IL-2.
本发明还提供另一种用于扩增活化淋巴细胞的培养系统, 其包括: The present invention also provides another culture system for amplifying activated lymphocytes, comprising:
(a)用于活化生物学样品中的淋巴细胞并同时进行第一个扩增步骤的 第一扩增容器, 其含有淋巴细胞活化剂和第一培养基, 所述第一培养基 含有高浓度的 IL-2; (a) a first amplification container for activating a lymphocyte in a biological sample and simultaneously performing a first amplification step, comprising a lymphocyte activator and a first medium, said first medium containing a high concentration IL-2;
(b)用于对活化淋巴细胞进行第二个扩增步骤的第二扩增容器, 其含 有第二培养基, 所述第二培养基含有低浓度的 IL-2; 和 (b) a second amplification vessel for performing a second amplification step on the activated lymphocytes, comprising a second medium comprising a low concentration of IL-2;
(c)用于对活化淋巴细胞进行第三个扩增步骤的第三扩增容器, 其含 有第三培养基, 所述第三培养基含有中等浓度的 IL-2。 (c) A third amplification vessel for performing a third amplification step on activated lymphocytes, comprising a third medium containing a medium concentration of IL-2.
优选地, 本发明的培养系统中使用的淋巴细胞活化剂是固定化的抗 CD3抗体。 Preferably, the lymphocyte activator used in the culture system of the present invention is an immobilized anti-CD3 antibody.
优选地, 在本发明的培养系统中, 所述第一培养基中的 IL-2的浓度 可以是例如 1000 U/ml至 6000 U/ml, 所述第二培养基中的 IL-2的浓度可 以是例如 1 U/ml至 500 U/ml, 所述第三培养基中的 IL-2的浓度可以是例 如 300 U/ml至 750 U/ml。 Preferably, in the culture system of the present invention, the concentration of IL-2 in the first medium may be, for example, 1000 U/ml to 6000 U/ml, and the concentration of IL-2 in the second medium. It may be, for example, 1 U/ml to 500 U/ml, and the concentration of IL-2 in the third medium may be, for example, 300 U/ml to 750 U/ml.
在本发明的培养系统中, 所述第一和 /或第二和 /或第三培养基中还可 以含有其他细胞因子, 例如 IL-7和 /或 IL-15。 优选地, 所述第一和 /或第 二和 /或第三培养基中 IL-7的浓度可以是例如 l-100ng/ml, 而 IL-15的浓 度可以是例如 l-200ng/ml。 In the culture system of the present invention, the first and/or second and/or third medium may further contain other cytokines such as IL-7 and/or IL-15. Preferably, said first and / or The concentration of IL-7 in the di- and/or third medium may be, for example, 1-100 ng/ml, and the concentration of IL-15 may be, for example, 1-200 ng/ml.
在本发明的培养系统中, 所述第一、 第二、 和第三培养基可以是相 同的或者不同的。 优选地, 所述第一培养基是 RPMI-1640培养基, 所述 第二培养基是 AIM-V培养基, 所述第三培养基是 DMEM培养基。 所述 第一、 第二、 和第三培养基也可以是混和培养基。 In the culture system of the present invention, the first, second, and third media may be the same or different. Preferably, the first medium is RPMI-1640 medium, the second medium is AIM-V medium, and the third medium is DMEM medium. The first, second, and third media may also be a mixed medium.
本发明还提供一种免疫治疗方法, 包括给个体施用通过上述本发明 的扩增活化淋巴细胞的方法而获得的扩增的活化淋巴细胞 (EAL)。 The present invention also provides an immunotherapy method comprising administering to an individual an expanded activated lymphocyte (EAL) obtained by the above method for amplifying activated lymphocytes of the present invention.
在本发明的免疫治疗方法的一个实施方式中, 通过上述扩增方法所 获得的淋巴细胞是扩增的活化自体淋巴细胞 (Expanded Activated Autologous Lymphocytes, g卩 EAAL), 且所述免疫治疗方法包括以下步 骤: In one embodiment of the immunotherapy method of the present invention, the lymphocytes obtained by the above amplification method are Expanded Activated Autologous Lymphocytes (g卩EAAL), and the immunotherapy method includes the following Steps:
(i)从个体获得含有淋巴细胞的生物学样品; (i) obtaining a biological sample containing lymphocytes from an individual;
(ii)采用上述本发明的扩增活化淋巴细胞的方法扩增所述淋巴细胞, 由此获得扩增的活化自体淋巴细胞; 和 (ii) amplifying the lymphocytes by the method of amplifying activated lymphocytes of the present invention described above, thereby obtaining amplified activated autologous lymphocytes;
(iii)将所述扩增的活化自体淋巴细胞回输给所述个体。 (iii) returning the amplified activated autologous lymphocytes to the individual.
本发明的免疫治疗方法可用于治疗患有例如肿瘤、 感染性疾病、 先 天性或后天性免疫缺陷症的个体。 可使用本发明的免疫治疗方法进行治 疗的肿瘤例如为肺癌、 贲门癌、 结肠癌、 乳腺癌、 髓母细胞瘤、 胃癌、 肾癌、 和恶性黑色素瘤, 且患有肿瘤的个体可以是正在接受放疗和 /或化 疗的个体。 可使用本发明的免疫治疗方法进行治疗的感染性疾病例如为 病毒感染, 特别是乙型和丙型肝炎病毒感染。 附图说明 The immunotherapeutic methods of the invention are useful for treating individuals suffering from, for example, tumors, infectious diseases, congenital or acquired immunodeficiency. Tumors that can be treated using the immunotherapeutic methods of the invention are, for example, lung cancer, cardiac cancer, colon cancer, breast cancer, medulloblastoma, gastric cancer, renal cancer, and malignant melanoma, and individuals with tumors may be receiving Individuals who receive radiation and/or chemotherapy. Infectious diseases which can be treated using the immunotherapy method of the present invention are, for example, viral infections, particularly hepatitis B and C virus infections. DRAWINGS
图 1 : 30例 2周活化 /扩增培养的细胞培养前后细胞扩增倍数分析。 图 2: 25例 2周活化 /扩增培养的细胞培养前后淋巴细胞表型分析。 图 3: EAAL回输后, 病人体内分泌 IFN- Y的外周血淋巴细胞比例 明显增多。 Figure 1: Analysis of cell expansion folds before and after cell culture in 30 cases of 2-week activation/amplification culture. Figure 2: Analysis of lymphocyte phenotype before and after cell culture in 25 cases of 2-week activation/amplification culture. Figure 3: After EAAL reinfusion, the proportion of peripheral blood lymphocytes secreting IFN-γ in patients is significantly increased.
图 4: EAAL回输后, 病人外周血中 CD8+ T细胞的比例明显升高 (p=0.0036)。 图 5: EAAL回输后, 病人外周血中 CD4+ T细胞和 CD8+ T细胞的 比例明显下降(p=0.0333)。 Figure 4: After EAAL reinfusion, the proportion of CD8+ T cells in peripheral blood of patients was significantly higher (p=0.0036). Figure 5: After EAAL reinfusion, the proportion of CD4+ T cells and CD8+ T cells in the peripheral blood of patients was significantly reduced (p=0.0333).
图 6: 复发的乳腺癌, 以本发明的 EAAL治疗后(5次回输)肿瘤标志 物水平下降。 具体实施方式 Figure 6: Recurrent breast cancer, with decreased levels of tumor markers after treatment with EAAL of the invention (5 reinfusions). detailed description
本发明提供了一种高效扩增活化淋巴细胞的方法和用于所述方法的 培养系统, 所述培养系统也称为高效扩增活化淋巴细胞的试剂盒 (Expanded Activated Lymphocyte Kit, EAL it) o 本发明的扩增方法基于传 统 CIK方法改良产生, 是一种高效 CIK细胞培养方法。 培养 /扩增方法 The present invention provides a method for efficiently amplifying activated lymphocytes and a culture system for the same, which is also called an Expanded Activated Lymphocyte Kit (EAL it) o The amplification method of the present invention is improved based on the conventional CIK method and is a highly efficient CIK cell culture method. Culture/amplification method
本发明的扩增活化淋巴细胞的方法包括- The method of amplifying activated lymphocytes of the present invention comprises -
(a)淋巴细胞活化步骤, 其中将含有淋巴细胞的生物学样品与淋巴细 胞活化剂相接触, 接触的时间和条件足以使得所述生物学样品中的淋巴 细胞发生活化; (a) a lymphocyte activation step, wherein the biological sample containing lymphocytes is contacted with a lymphocyte activator for a time and conditions sufficient to activate lymphocytes in the biological sample;
(b)第一个扩增步骤, 其中将步骤 (a)中得到的活化淋巴细胞在含有高 浓度 IL-2的第一培养基中进行培养; (b) a first amplification step in which the activated lymphocytes obtained in the step (a) are cultured in a first medium containing a high concentration of IL-2;
(c)第二个扩增步骤, 其中将步骤 (b)中得到的淋巴细胞在含有低浓度 IL-2的第二培养基中进行培养; (c) a second amplification step, wherein the lymphocytes obtained in the step (b) are cultured in a second medium containing a low concentration of IL-2;
(d)第三个扩增步骤, 其中将步骤 (c)中得到的淋巴细胞在含有中等浓 度 IL-2的第三培养基中进行培养; 和 (d) a third amplification step in which the lymphocytes obtained in the step (c) are cultured in a third medium containing a medium concentration of IL-2;
(e)收集步骤 (d)中得到的扩增的活化淋巴细胞。 (e) Collecting the amplified activated lymphocytes obtained in the step (d).
在本发明的方法的一个实施方式中, 淋巴细胞活化步骤与第一个扩 增步骤在同一个培养容器中同时进行。 淋巴细胞 In one embodiment of the method of the invention, the lymphocyte activation step and the first amplification step are performed simultaneously in the same culture vessel. Lymphocyte
能够使用本发明的方法进行培养和扩增的淋巴细胞可来自外周血淋 巴细胞。 待扩增的淋巴细胞也可以是其他生物学样品中存在的淋巴细 胞, 例如上皮淋巴细胞、 肿瘤内浸润淋巴细胞、 癌性腹水或胸水中的浸 润淋巴细胞等。 由于外周血中的淋巴细胞分离简便、 分离后淋巴细胞生 存率高等原因, 因此优选地使用分离自新鲜外周血的淋巴细胞。 淋巴细胞活化 Lymphocytes that can be cultured and expanded using the methods of the invention can be derived from peripheral blood lymphocytes. The lymphocytes to be expanded may also be lymphocytes present in other biological samples, such as epithelial lymphocytes, intratumoral infiltrating lymphocytes, cancerous ascites or pleural immersion. Run lymphocytes and so on. Lymphocytes isolated from fresh peripheral blood are preferably used because of the ease of separation of lymphocytes in peripheral blood, high lymphocyte survival rate after isolation, and the like. Lymphocyte activation
术语 "淋巴细胞活化"是指应用特异性抗原或非特异性淋巴细胞刺 激剂 /分子刺激淋巴细胞, 导致淋巴细胞合成大分子物质 (RNA、 蛋白 质、 DNA)及细胞因子。 淋巴细胞活化后进行细胞增殖, 并分化为不同功 能的子代效应细胞和记忆细胞。 The term "lymphocyte activation" refers to the use of specific antigens or non-specific lymphocyte stimulators/molecules to stimulate lymphocytes, resulting in the synthesis of macromolecular substances (RNA, protein, DNA) and cytokines by lymphocytes. After lymphocyte activation, the cells proliferate and differentiate into progeny effector cells and memory cells of different functions.
术语 "淋巴细胞活化剂" 是指能够刺激淋巴细胞活化的试剂 /分子。 例如, 可以使用抗 CD3抗体作为淋巴细胞活化剂, 该试剂刺激 T细胞表 面的 CD3分子, 通过 T细胞受体有效活化 T淋巴细胞。 淋巴细胞有丝分 裂原是另一类有效的淋巴细胞活化剂。 这类分子快速活化淋巴细胞, 其 作用不通过 T淋巴细胞受体。 The term "lymphocyte activator" refers to a reagent/molecule that is capable of stimulating lymphocyte activation. For example, an anti-CD3 antibody can be used as a lymphocyte activating agent which stimulates CD3 molecules on the surface of T cells and efficiently activates T lymphocytes through T cell receptors. Lymphocyte mitogens are another class of potent lymphocyte activators. These molecules rapidly activate lymphocytes and do not pass through T lymphocyte receptors.
合适的淋巴细胞活化剂的实例是抗 CD3抗体。 在本发明的淋巴细胞 扩增方法的一个具体实施方式中, 所述淋巴细胞活化剂是固定化于基质 表面的抗 CD3抗体。 使用固定化的抗 CD3抗体是优选的, 因为其主要活 化具有细胞杀伤功能的 CD8+ T细胞, 这特别有利于提高所扩增的淋巴细 胞的抗肿瘤活性。 制备含有固定化的抗 CD3抗体的培养容器的方法是己 知的, 且在下文中有详细的描述。 细胞因子 An example of a suitable lymphocyte activator is an anti-CD3 antibody. In a specific embodiment of the lymphocyte expansion method of the present invention, the lymphocyte activating agent is an anti-CD3 antibody immobilized on a surface of a substrate. The use of an immobilized anti-CD3 antibody is preferred because it mainly activates CD8+ T cells having a cell killing function, which is particularly advantageous for enhancing the antitumor activity of the expanded lymphocytes. Methods of preparing culture vessels containing immobilized anti-CD3 antibodies are known and are described in detail below. Cytokine
传统淋巴细胞扩增方法所采用的促进淋巴细胞增殖的细胞因子主要 为 IL-2、 IL-4、 IL-l a及 γ干扰素(IFN- Y )。 这 细胞因子的配伍形式主 要使得 CD3+CD56+细胞、 CD4+ T细胞和 CD8+ T细胞增殖。 The cytokines used to promote lymphocyte proliferation by the conventional lymphocyte expansion method are mainly IL-2, IL-4, IL-l and IFN-γ. The compatible form of this cytokine primarily causes proliferation of CD3+CD56+ cells, CD4+ T cells, and CD8+ T cells.
在本发明中, 细胞因子主要使用 IL-2。 IL-2可以高效率、 高专一性 地扩增 CD8+ T细胞, 而 CD8+ T细胞是免疫系统发挥抗肿瘤作用的最重 要的细胞成分。 传统淋巴细胞扩增方法所采用的细胞因子 IL-2的浓度从 培养开始至结束, 始终使用同一浓度。 而与传统方法不同的是, 本发明 在不同培养阶段使用不同浓度的 IL-2。 In the present invention, cytokines mainly use IL-2. IL-2 can amplify CD8+ T cells with high efficiency and specificity, and CD8+ T cells are the most important cellular components of the immune system to exert anti-tumor effects. The concentration of the cytokine IL-2 used in the conventional lymphocyte expansion method is always the same concentration from the start to the end of the culture. Unlike the conventional method, the present invention uses different concentrations of IL-2 in different culture stages.
根据本发明, 为在短时间内获得大量具有迅速增殖能力的淋巴细 胞, 在第一个扩增步骤 (b)中将步骤 (a)中得到的活化淋巴细胞在含有高浓 度 IL-2的第一培养基中进行培养。 优选地, 所述第一培养基中 IL-2的浓 度范围是例如 1000 U/ml至 6000 U/ml。 细胞在所述第一个扩增步骤中生 长至第 7-14天。 考虑到培养液营养的消耗及 IL-2和其它细胞因子活性的 降低, 每天适量地添加新鲜培养液是非常重要的。 在第二个扩增步骤 (c) 中, 将步骤 (b)中得到的淋巴细胞在含有低浓度 IL-2的第二培养基中继续 培养, 这样可使得细胞在培养容器中保持增殖速度, 并不被过度活化。 优选地, 所述第二培养基中 IL-2的浓度范围是例如 1 U/ml至 500 U/ml。 细胞在所述第二个扩增步骤中生长 3-7天。 培养的第 10 - 14天进入第三 个扩增步骤 (d), 其中将步骤 (c)中得到的淋巴细胞在含有中等浓度 IL-2的 第三培养基中进行培养。 优选地, 所述第三培养基中 IL-2的浓度范围是 例如 300U/ml至 750 U/ml。 细胞在所述第三个扩增步骤中继续培养 3 - 7 天, 以便大量扩增细胞, 使细胞数目满足临床使用要求。 采用本发明的 方法, 淋巴细胞可达到 100倍、 甚至 1000倍以上的扩增。 According to the present invention, in order to obtain a large amount of lymphocytes having rapid proliferative ability in a short time The activated lymphocytes obtained in the step (a) are cultured in the first medium containing the high concentration of IL-2 in the first amplification step (b). Preferably, the concentration of IL-2 in the first medium ranges, for example, from 1000 U/ml to 6000 U/ml. The cells were grown to the 7-14th day in the first amplification step. Considering the nutrient consumption of the culture medium and the decrease in the activity of IL-2 and other cytokines, it is very important to add a proper amount of fresh culture solution every day. In the second amplification step (c), the lymphocytes obtained in the step (b) are further cultured in a second medium containing a low concentration of IL-2, so that the cells maintain the proliferation rate in the culture vessel. Not overactivated. Preferably, the concentration of IL-2 in the second medium ranges, for example, from 1 U/ml to 500 U/ml. The cells are grown for 3-7 days in the second amplification step. The 10th-14th day of the culture enters a third amplification step (d) in which the lymphocytes obtained in the step (c) are cultured in a third medium containing a medium concentration of IL-2. Preferably, the concentration of IL-2 in the third medium ranges, for example, from 300 U/ml to 750 U/ml. The cells are further cultured for 3 - 7 days in the third amplification step to extensively expand the cells to meet the clinical use requirements. With the method of the present invention, lymphocytes can be amplified by a factor of 100 or more.
除 IL-2 以外, 本发明的扩增方法中还可进一步加入其他能够促进淋 巴细胞增殖的细胞因子。 例如, 还可在培养基中加入白细胞介素 -7 (IL-7) 和 /或白细胞介素 -15 (IL-15), 以便通过维持记忆细胞的生存能力并减少细 胞活化后的死亡, 从而达到最好的细胞扩增效果和细胞活性的保持。 在 本发明的方法中, 所述步骤 (b)和 /或 (c)和 /或 (d)的培养基中 IL-7的浓度优 选地是 l-100ng/ml, 而 IL-15的浓度优选地是 l-200ng/ml。 培养基 In addition to IL-2, other cytokines capable of promoting proliferation of lymphocytes may be further added to the amplification method of the present invention. For example, interleukin-7 (IL-7) and/or interleukin-15 (IL-15) can also be added to the culture medium to maintain memory cell viability and reduce death after cell activation. Achieve the best cell expansion and maintenance of cell viability. In the method of the present invention, the concentration of IL-7 in the medium of the step (b) and/or (c) and/or (d) is preferably from 1 to 100 ng/ml, and the concentration of IL-15 is preferably The ground is l-200ng/ml. Medium
传统方法所采用的淋巴细胞培养 /扩增方案均采用一种固定的细胞培 养基, 如 RPMI-1640或 DMEM等。 但在本发明的方法中, 所述步骤 (b)、 (c), 和 (d)的培养基可以是不同的。 优选地, 所述第一培养基是 RPMI-1640培养基, 所述第二培养基是 AIM-V培养基, 所述第三培养基 是 DMEM培养基。 通过改变不同细胞培养阶段所釆用的细胞培养基, 可 以为细胞提供更为充足的营养成分, 提高细胞的增殖能力, 进而增加其 扩增倍数。 此外, 在本发明的方法中, 所述第一、 第二、 和第三培养基 也可以是混和培养基。 此外, 所述第一、 第二、 和第三培养基还可以加 入其他有助于淋巴细胞扩增的成分, 例如胎牛血清或者人血清。 培养基 中血清的浓度为 1 - 20%。 使用前将血清在 37Ό解冻后, 在 56°C加热 30 分钟使其灭活。 The lymphocyte culture/amplification protocol used in the conventional method employs a fixed cell culture medium such as RPMI-1640 or DMEM. However, in the method of the present invention, the medium of the steps (b), (c), and (d) may be different. Preferably, the first medium is RPMI-1640 medium, the second medium is AIM-V medium, and the third medium is DMEM medium. By changing the cell culture medium used in different cell culture stages, it is possible to provide more abundant nutrients to the cells, increase the cell's proliferative capacity, and increase the amplification factor. Further, in the method of the present invention, the first, second, and third medium may also be a mixed medium. In addition, the first, second, and third media may also be added Into other components that help lymphocyte expansion, such as fetal bovine serum or human serum. The concentration of serum in the medium is 1 - 20%. The serum was thawed at 37 °C before use and inactivated by heating at 56 ° C for 30 minutes.
术语 "混和培养基"在此是指常规培养基, 如 RPMI-1640, DMEM, IMEM,AIM-V等, 的一种或多种的混合。 培养方式 The term "mixed medium" as used herein refers to a mixture of one or more of conventional media such as RPMI-1640, DMEM, IMEM, AIM-V, and the like. Training method
在本发明的方法中, 可以使用任何适合用于淋巴细胞活化和扩增的 培养容器。 例如, 本发明的方法可采用细胞培养瓶进行细胞培养, 也可 采用密闭的细胞培养袋系统进行细胞的扩增培养。 培养容器的容积应该 满足对细胞进行大量扩增的需要。 可用于本发明的培养袋例如可以容纳 相当于 10-20个 T225细胞培养瓶所容纳的细胞数目, 这样可大大降低培 养细胞的工作量, 同时密闭系统大幅度降低了操作中污染的可能性。 例 如, 可以通过瓶式扩增进行所述的第一个扩增步骤, 并通过袋式扩增进 行所述的第二个扩增步骤和所述的第三个扩增步骤。 优选地, 在本发明 的方法中, 淋巴细胞活化步骤与第一个扩增步骤在同一个瓶式扩增容器 中同时进行。 In the method of the present invention, any culture vessel suitable for lymphocyte activation and amplification can be used. For example, the method of the present invention may employ a cell culture flask for cell culture, or a closed cell culture bag system for cell expansion and culture. The volume of the culture vessel should meet the need for extensive expansion of the cells. The culture bag which can be used in the present invention can, for example, accommodate the number of cells accommodated in 10-20 T225 cell culture flasks, which greatly reduces the workload of the cultured cells, while the closed system greatly reduces the possibility of contamination during the operation. For example, the first amplification step can be performed by bottle amplification, and the second amplification step and the third amplification step can be performed by bag amplification. Preferably, in the method of the invention, the lymphocyte activation step and the first amplification step are performed simultaneously in the same vial amplification vessel.
优选地, 所述的第一个扩增步骤进行 7-14天, 所述的第二个扩增步 骤进行 3至 7天, 且所述的第三个扩增步骤进行 3至 7天。 在本发明的方 法中, 在进行步骤 (e)之前, 可以将第三个扩增步骤重复一或多次。 使用本发明的扩增活化淋巴细胞的方法, 所述生物学样品中的淋巴 细胞可被扩增至少 10倍、 优选地至少 100倍、 更优选地至少 1000倍。 优选地, 其中扩增后的细胞主要为 CD8+ T细胞和 NK细胞, 例如, 所扩 增的活化淋巴细胞中, CD8+ T细胞可 >50%。 培养 /扩增系统 Preferably, the first amplification step is performed for 7-14 days, the second amplification step is performed for 3 to 7 days, and the third amplification step is performed for 3 to 7 days. In the method of the present invention, the third amplification step may be repeated one or more times before performing step (e). Using the method of amplifying activated lymphocytes of the present invention, lymphocytes in the biological sample can be amplified at least 10 fold, preferably at least 100 fold, more preferably at least 1000 fold. Preferably, wherein the expanded cells are mainly CD8+ T cells and NK cells, for example, CD8+ T cells can be >50% in the expanded activated lymphocytes. Culture/amplification system
制备活化培养容器 Preparation of activated culture vessel
活化培养容器用于活化生物学样品中的淋巴细胞, 其含有淋巴细胞 活化剂。 在一个具体的实施方式中, 本发明的培养 /扩增系统使用的活化培养 容器含有固定化于不溶性载体例如所述容器表面的抗 CD3抗体作为淋巴 细胞活化剂。 The activation culture vessel is used to activate lymphocytes in a biological sample containing a lymphocyte activator. In a specific embodiment, the activation culture vessel used in the culture/amplification system of the present invention contains an anti-CD3 antibody immobilized on an insoluble carrier such as the surface of the container as a lymphocyte activating agent.
在制备含有固定化于不溶性载体的抗 CD3.抗体的活化培养容器时, 可将抗 CD3抗体以灭菌的磷酸氯化钠缓冲液(PBS)稀释成 1 - 50 w g/ml 的浓度使用。 不足 1 μ g/ml的浓度时, T细胞的增殖倾向于不充分, 超过 50 y g/ml时, T细胞的增殖虽然充分, 但没有固定于不溶性载体的、 自 由的抗 CD3抗体倾向于增多。 另外, 作为稀释液, 只要是灭菌的磷酸氯 化钠缓冲液等生理溶液即可, 不作特别限定, 作为保护剂, 可加入白蛋 白、 琼脂糖等。 In the preparation of an activated culture vessel containing an anti-CD3. antibody immobilized on an insoluble carrier, the anti-CD3 antibody can be diluted with a sterile sodium phosphate buffer (PBS) to a concentration of 1 - 50 w g/ml. When the concentration is less than 1 μg/ml, the proliferation of T cells tends to be insufficient. When the proliferation of T cells exceeds 50 μg/ml, the proliferation of T cells is not sufficient, but the free anti-CD3 antibody which is not immobilized on the insoluble carrier tends to increase. In addition, the diluent is not particularly limited as long as it is a physiological solution such as a sterilized sodium phosphate buffer solution, and as a protective agent, white protein, agarose or the like may be added.
将稀释的抗 CD3抗体无菌地平铺于培养瓶的底面。 固定时的反应温 度为 4-40°C, 更优选为 4-25V , 最优选为 4-10°C。 不足 4°C时, 抗 CD3 抗体的固定化效率倾向于降低, 超过 40°C时, 抗 CD3抗体的活性倾向于 降低。 固定化时的反应时间为 30分钟 -24小时, 优选 1小时至 24小时。 不足 30分钟时, 抗 CD3抗体的固定化量倾向于降低, 超过 24小时时, 固定时间过长, 生产效率倾向于降低。 The diluted anti-CD3 antibody was aseptically plated on the bottom surface of the culture flask. The reaction temperature at the time of fixation is 4 to 40 ° C, more preferably 4 to 25 V, and most preferably 4 to 10 ° C. At less than 4 °C, the immobilization efficiency of the anti-CD3 antibody tends to decrease, and when it exceeds 40 °C, the activity of the anti-CD3 antibody tends to decrease. The reaction time in the immobilization is from 30 minutes to 24 hours, preferably from 1 hour to 24 hours. At less than 30 minutes, the immobilization amount of the anti-CD3 antibody tends to decrease, and when it exceeds 24 hours, the fixation time is too long, and the production efficiency tends to decrease.
紧接着上述抗 CD3抗体的固定化, 要除去没被固定的残余的抗 CD3 抗体稀释液。 通过使用磷酸氯化钠缓冲液(PBS)等适当的清洗液清洗没 有被固定的抗 CD3抗体, 使其干燥。 干燥方法可以在去除清洗液以后利 用 4-10°C冷库中的自然干燥、 10-40°C的加温干燥、 冻干等方法。 考虑到 抗 CD3抗体的活性和稳定性, 优选冻干, 最优选 4-10°C冷库中的自然干 燥。 如上所述, 在不溶性载体的表面抗 CD3抗体被固定化, 得到该抗 CD3抗体的固定化表面被干燥的 T细胞培养用载体, 无菌密封后获得活 化培养容器。 制备第一扩增容器 Immediately following the immobilization of the above anti-CD3 antibody, the residual anti-CD3 antibody dilution which was not immobilized was removed. The anti-CD3 antibody which has not been immobilized is washed with an appropriate washing solution such as sodium phosphate buffer (PBS) and dried. The drying method can be carried out by natural drying in a 4-10 ° C cold storage, heating drying at 10-40 ° C, lyophilization, etc. after removing the cleaning liquid. In view of the activity and stability of the anti-CD3 antibody, lyophilization is preferred, and natural drying in a 4-10 °C cold storage is most preferred. As described above, the anti-CD3 antibody is immobilized on the surface of the insoluble carrier, and a carrier for T cell culture in which the immobilized surface of the anti-CD3 antibody is dried is obtained, and the culture vessel is obtained aseptically sealed. Preparation of the first amplification container
第一扩增容器用于对活化淋巴细胞进行第一个扩增步骤, 其含有第 一培养基, 所述第一培养基含有高浓度的 IL-2。 优选地, 所述第一培养 基中的 IL-2的浓度是 1000 U/ml至 6000 U/ml。 The first amplification vessel is used to perform a first amplification step on the activated lymphocytes, which contains a first medium containing a high concentration of IL-2. Preferably, the concentration of IL-2 in the first medium is from 1000 U/ml to 6000 U/ml.
用基础培养基 RPMI-1640培养基稀释 IL-2浓度为 1000- 6000 U/ml。 为提高细胞的存活率和生存时间, 可加入 IL-7和 /或 IL-15。 浓度分别为 l-100ng/ml和 l-200ng/ml。 随后, 将培养基配成体积比为含有 1 - 20%的 胎牛血清或人血清(将血清在 37°C解冻后, 在 56°C加热 30分钟使其灭活) 的细胞培养基, 但为了促进淋巴细胞增殖, 一般优选 2-10(V/V)%。 培养 基优选地使用灭菌的 RPMI-1640培养基, 但并不仅限于此, 其他适合于 淋巴细胞的培养基或混合培养基均可使用。 如此制备得到用于第一扩增 容器的第一培养基。 The IL-2 concentration was diluted 1000-6000 U/ml with basal medium RPMI-1640 medium. To increase cell viability and survival time, IL-7 and/or IL-15 can be added. The concentrations were 1-100 ng/ml and 1-200 ng/ml, respectively. Subsequently, the medium is formulated into a cell culture medium having a volume ratio of 1 - 20% fetal calf serum or human serum (the serum is thawed at 37 ° C and heated at 56 ° C for 30 minutes), but In order to promote lymphocyte proliferation, it is generally preferably 2 to 10 (V/V)%. The culture medium is preferably a sterilized RPMI-1640 medium, but is not limited thereto, and other medium suitable for lymphocytes or a mixed medium can be used. The first medium for the first amplification vessel is thus prepared.
当活化步骤与第一扩增步骤同时进行时, , 可直接将如上所述配制 的培养基加入到上述活化培养容器中。 制备第二扩增容器 When the activation step is performed simultaneously with the first amplification step, the medium prepared as described above can be directly added to the above-described activated culture vessel. Preparation of a second amplification container
第二扩增容器用于对活化淋巴细胞进行第二个扩增步骤, 其含有第 二培荞基, 所述第二培养基含有低浓度的 IL-2。 优选地, 所述第二培养 基中的 IL-2的浓度是 1 U/ml至 500 U/ml。 优选地, 第二扩增容器可以使 用袋式培养容器。 The second amplification vessel is for performing a second amplification step on the activated lymphocytes, which contains a second culture medium, and the second medium contains a low concentration of IL-2. Preferably, the concentration of IL-2 in the second medium is from 1 U/ml to 500 U/ml. Preferably, the second expansion vessel may use a bag culture vessel.
用 AIM-V培养基稀释 IL-2浓度为 1 - 500 U/ml。 为提高细胞的存活 率和生存时间, 可加入 IL-7和 /或 IL-15 o 浓度分别为 l-100ng/ml和 1- 200ng/ml。 随后, 将培养基配成体积比为含有 1 - 20%的胎牛血清(将血 清在 37°C解冻后, 在 56°C加热 30分钟使其灭活)的细胞培养基。 如此制 备得到用于第二扩增容器的第二培养基。 第二培养培养基采用 AIM-V培 养基效果最好, 但并不仅限于此, 其它适合于淋巴细胞的培养基或混合 培养基亦可。 制备第三扩增容器 The IL-2 concentration was diluted 1 - 500 U/ml with AIM-V medium. To increase cell viability and survival time, IL-7 and / or IL-15 o concentrations of 1-100 ng / ml and 1- 200 ng / ml can be added. Subsequently, the medium was formulated into a cell culture medium having a volume ratio of 1 - 20% fetal calf serum (the serum was thawed at 37 ° C and heated at 56 ° C for 30 minutes). A second medium for the second amplification vessel is thus prepared. The second culture medium preferably uses AIM-V medium, but is not limited thereto, and other medium or mixed medium suitable for lymphocytes may also be used. Preparation of a third amplification container
第三扩增容器用于对活化淋巴细胞进行第三个扩增步骤, 其含有第 三培养基, 所述第三培养基含有中等浓度的 IL-2。 优选地, 所述第三培 养基中的 IL-2的浓度是 300 U/ml至 750 U/ml。 优选地, 第三扩增容器可 以使用袋式培养容器。 The third amplification vessel is used to perform a third amplification step on the activated lymphocytes, which contains a third medium containing a medium concentration of IL-2. Preferably, the concentration of IL-2 in the third medium is from 300 U/ml to 750 U/ml. Preferably, the third amplification container can use a bag culture container.
用 DMEM培养基稀释 IL-2浓度为 300 - 750 U/ml。 为提高细胞的存 活率和生存时间, 可加入 IL-7和 /或 IL-15, 浓度分别为 l-100ng/ml和 1- 200ng/ml。 随后, 将培养基配成体积比为含有 1 - 20%的胎牛血清(将血 清在 37°C解冻后, 在 56°C加热 30分钟使其灭活)的细胞培养基。 如此制 备得到用于第三扩增容器的第三培养基。 第三培养基采用 DMEM培养基 效果最好, 但并不仅限于此, 其它适合于淋巴细胞的培养基或混合培养 基亦可。 免疫治疗方法 The IL-2 concentration was diluted from DMEM medium to 300 - 750 U/ml. In order to improve cell survival and survival time, IL-7 and / or IL-15 can be added at concentrations of 1-100 ng/ml and 1- 200 ng/ml. Subsequently, the medium was formulated into a cell culture medium in a volume ratio of 1 - 20% fetal calf serum (the serum was thawed at 37 ° C and heated at 56 ° C for 30 minutes). The third medium for the third amplification vessel was thus prepared. The third medium is best used in DMEM medium, but it is not limited thereto, and other medium suitable for lymphocytes or mixed medium may also be used. Immunotherapy
本发明还提供了一种免疫治疗方法, 包括给个体施用通过上述本发 明的扩增活化淋巴细胞的方法而获得的扩增的活化淋巴细胞 (EAL)。 The present invention also provides an immunotherapy method comprising administering to an individual an expanded activated lymphocyte (EAL) obtained by the method of amplifying activated lymphocytes of the present invention described above.
在本发明的免疫治疗方法的一个实施方式中, 通过上述扩增方法所 获得的淋巴细胞是扩增的活化自体淋巴细胞 (Expanded Activated Autologous Lymphocytes , gp EAAL), 且所述免疫治疗方法包括以下步 骤: In one embodiment of the immunotherapy method of the present invention, the lymphocytes obtained by the above amplification method are Expanded Activated Autologous Lymphocytes (gp EAAL), and the immunotherapy method comprises the following steps :
(i)从个体获得含有淋巴细胞的生物学样品; (i) obtaining a biological sample containing lymphocytes from an individual;
(ii)采用上述本发明的扩增活化淋巴细胞的方法扩增所述淋巴细胞, 由此获得扩增的活化自体淋巴细胞; 和 (ii) amplifying the lymphocytes by the method of amplifying activated lymphocytes of the present invention described above, thereby obtaining amplified activated autologous lymphocytes;
(iii)将所述扩增的活化自体淋巴细胞回输给所述个体。 (iii) returning the amplified activated autologous lymphocytes to the individual.
在本发明的免疫治疗方法中, 当所使用的扩增的淋巴细胞是自体淋 巴细胞时, 我们称其为扩增活化自体淋巴细胞疗法 (即 Expanded Activated Autologous Lymphocytes (EAAL) Therapy)。 在治疗中使用的淋 巴细胞来源于病人自身的少量外周血。 这些淋巴细胞在约 2至 4周的短 时间内, 可以活化扩增达 100 - 1000倍, 再回输于病人体内。 本发明的免 疫治疗方法可用于治疗患有例如肿瘤、 感染性疾病、 先天性或后天性免 疫缺陷症、 以及器官移植后感染性疾病及感染诱发的肿瘤的个体。 In the immunotherapy method of the present invention, when the amplified lymphocytes used are autologous lymphocytes, we call them Expanded Activated Autologous Lymphocytes (EAAL) Therapy. The lymphocytes used in the treatment are derived from a small amount of peripheral blood of the patient. These lymphocytes can be activated and amplified 100-1000 times in a short period of about 2 to 4 weeks and then returned to the patient. The immunotherapy method of the present invention can be used for treating an individual suffering from, for example, a tumor, an infectious disease, a congenital or acquired immunodeficiency defect, and an infectious disease caused by an organ transplant and an infection-induced tumor.
与传统 CIK细胞不同, 用此方法获得的活化细胞大部分是 CD8+ T 淋巴细胞(>50%)。 CD8+ T淋巴细胞在杀伤病毒感染的细胞和癌症细胞 中发挥重要的功能。 人体内 T淋巴细胞包括许多针对不同抗原的特异性 细胞。 这些细胞经过培养后, 理论上它们会针对不同的肿瘤抗原 /病毒抗 原具有广泛的识别和杀伤效果。 治疗的目的, 是将这样的淋巴细胞在体 外活化增殖, 提高其功能, 在治疗中使用, 以杀伤肿瘤细胞。 可使用本发明的免疫治疗方法进行治疗的肿瘤例如为肺癌、 贲门 癌、 结肠癌、 乳腺癌、 髓母细胞瘤、 胃癌、 肾癌、 和恶性黑色素瘤, 且 患有肿瘤的个体可以是正在接受放疗和 /或化疗的个体。 Unlike conventional CIK cells, the activated cells obtained by this method are mostly CD8+ T lymphocytes (>50%). CD8+ T lymphocytes play an important role in killing virus-infected cells and cancer cells. Human T lymphocytes include many specific cells directed against different antigens. These cells, after culturing, theoretically have broad recognition and killing effects against different tumor antigens/viral antigens. The purpose of treatment is to activate and proliferate such lymphocytes in vitro to enhance their function and to be used in therapy to kill tumor cells. Tumors that can be treated using the immunotherapeutic methods of the invention are, for example, lung cancer, cardiac cancer, colon cancer, breast cancer, medulloblastoma, gastric cancer, renal cancer, and malignant melanoma, and individuals with tumors may be receiving Individuals who receive radiation and/or chemotherapy.
对一部分慢性病毒感染的病人, 如乙型和丙型肝炎病人, 该方法还 可有效清除感染的病毒。 因此也可使用本发明的免疫治疗方法治疗感染 性疾病, 例如病毒感染, 特别是乙型肝炎病毒感染和丙型肝炎病毒感 染。 本发明的主要优势在于: For some patients with chronic viral infections, such as hepatitis B and C, this method can also effectively remove infected virus. Therefore, the immunotherapy method of the present invention can also be used to treat infectious diseases such as viral infections, particularly hepatitis B virus infection and hepatitis C virus infection. The main advantages of the invention are:
• 培养系统可为标准化的试剂盒系统; • The culture system can be a standardized kit system;
• 仅需采集 20-50ml供者或者自身外周血用于分离淋巴细胞; • 对供者或病人淋巴系统损伤小; • Only need to collect 20-50ml donor or self-peripheral blood for lymphocyte separation; • Little damage to the donor or patient lymphatic system;
• 节约费用; • Saving costs;
• 提高病人对放化疗的耐受, 可与放化疗联合治疗; • Improve patient tolerance to chemoradiotherapy, combined with radiotherapy and chemotherapy;
• 可高频率治疗; • High frequency treatment;
• 扩增效率高, 可达 100— 1000倍细胞扩增; • High amplification efficiency, up to 100-1000 times cell expansion;
• 自体不同时间、 不同个体之间、 不同实验室间均具有稳定的可重 复性; • Stable reproducibility at different times, between individuals, and between laboratories;
• 细胞稳定, 在扩增后 24小时内保持细胞活性, 可在 12小时内细 胞回输; • The cells are stable, maintain cell viability within 24 hours after amplification, and can be returned within 12 hours;
• 可长途运输。 实施例 • Long distance transportation. Example
以下实施例用于例证本发明, 其无意于以任何方式对本发明进行限 制。 The following examples are intended to illustrate the invention and are not intended to limit the invention in any way.
1.细胞活化: 1. Cell activation:
首先制备以固定化的抗 CD3 抗体包被的培养瓶。 将抗 CD3抗体 (Ortho)以灭菌的磷酸氯化钠缓冲液(PBS)稀释至 10 w g/ml。 将稀释的抗 CD3抗体无菌地平铺于培养瓶(Costa)的底面。 4°C过夜。 用磷酸氯化钠 缓冲液(PBS)清洗没有被固定的抗 CD3抗体后在 4-10°C冷库中自然干 燥, 得到该抗 CD3抗体的固定化表面被干燥的 T细胞培养用载体, 无菌 密封后获得活化培养容器。 First, a culture flask coated with an immobilized anti-CD3 antibody was prepared. Anti-CD3 antibody (Ortho) was diluted to 10 wg/ml in sterile sodium phosphate buffer (PBS). The diluted anti-CD3 antibody was aseptically plated on the bottom of a culture flask (Costa). 4 ° C overnight. Wash the anti-CD3 antibody without immobilization with sodium phosphate buffer (PBS) and dry naturally in a cold storage at 4-10 °C. After drying, the immobilized surface of the anti-CD3 antibody was dried, and the carrier for T cell culture was aseptically sealed to obtain an activated culture vessel.
用肝素盐水处理的注射器采集人静脉血全血 20ml, 加入 20ml生理 盐水, 进行 2倍稀释, 将每 10ml稀释后的血液加在 3ml的密度梯度离心 剂 Ficoll-Paque (Sigma- Aldrich Corp., St. Louis, MO, U.S.A.)上层, 在 1600rpm、 25 分钟、 20 °C条件下进行离心 (Beckman, Fullerton, CA, U.S.A., S4750A), 回收淋巴细胞。 将分离的淋巴细胞用 RPMI-1640培养 基 (Invitrogen, Carlsbad, CA, U.S.A.)洗 3次, 最后制成细胞密度约 2X 106/ml的细胞悬液。 将此细胞悬浊液 5ml (1 X 107个)加入上述活化培养容 器(包被了固定化的抗 CD3抗体的培养瓶)中, 在 50ml含 6000U/ml的 IL-2的如上所述的第一扩增试剂中开始培养。 Human hemorrhage whole blood 20 ml was collected with a heparin-treated syringe, 20 ml of physiological saline was added, and 2-fold dilution was performed. Each 10 ml of diluted blood was added to a 3 ml density gradient centrifuge Ficoll-Paque (Sigma-Aldrich Corp., St . Louis, MO, USA) Upper layer, centrifuged at 1600 rpm, 25 minutes, 20 °C (Beckman, Fullerton, CA, USA, S4750A) to recover lymphocytes. The isolated lymphocytes were washed 3 times with RPMI-1640 medium (Invitrogen, Carlsbad, CA, USA), and finally a cell suspension having a cell density of about 2×10 6 /ml was prepared. 5 ml of this cell suspension (1×10 7 ) was added to the above activated culture vessel (culture flask coated with immobilized anti-CD3 antibody) in 50 ml of IL-2 containing 6000 U/ml as described above. The culture is started in the first amplification reagent.
2. 细胞扩增- 培养的第 2天起, 显微镜(中国重庆光学仪器厂, CKX-41)下观察细 胞, 如细胞增殖良好, 共加入 180ml如上所述的第一扩增试剂, 细胞在 此条件下生长至第 7天。 考虑到培养液营养的消耗及 IL-2 (美国诺华制药 厂, 中国北京四环制药厂)和其它细胞因子活性的降低, 每天适量地添加 新鲜培养液是非常重要的。 2. Cell expansion - On the second day of culture, observe the cells under the microscope (Chongqing Optical Instrument Factory, CKX-41). If the cells proliferate well, add 180ml of the first amplification reagent as described above. Growth was carried out until day 7 under conditions. Considering the nutrient consumption of the culture medium and the reduction of IL-2 (Nova Chemical Factory, Beijing Sihuan Pharmaceutical Factory, China) and other cytokine activities, it is very important to add a proper amount of fresh culture solution every day.
将细胞及培养基一起, 在培养的第 7天后转入如上所述的第二扩增 试剂中, 在细胞培养袋中继续生长 3天。 The cells and the medium were transferred to the second amplification reagent as described above after the 7th day of the culture, and growth was continued for 3 days in the cell culture bag.
培养的第 10天, 将如上所述的第二扩增试剂中的细胞与如上所述的 第三扩增试剂混合均匀后, 在两个细胞培养袋中继续培养 4天。 如需更 多数目的细胞, 可以继续将两个细胞培养袋中的细胞再与一个如上所述 的第三扩增试剂中的试剂混合均匀后, 在三个培养袋中继续培养。 On the 10th day of the culture, the cells in the second amplification reagent as described above were uniformly mixed with the third amplification reagent as described above, and then cultured in the two cell culture bags for 4 days. If a larger number of cells are required, the cells in the two cell culture bags can be further mixed with a reagent in the third amplification reagent as described above, and the culture is continued in three culture bags.
3. 细胞收获: 3. Cell harvesting:
将培养袋中的细胞转入离心管中, 1600rpm, 离心 10分钟。 细胞用 1%白蛋白 (中国北京天坛生物制品公司)溶液洗两次后, 重悬于 5%的白 蛋白-生理盐水中, 进行以下检测。 The cells in the culture bag were transferred to a centrifuge tube, centrifuged at 1600 rpm for 10 minutes. The cells were washed twice with a solution of 1% albumin (Beijing Tiantan Biological Products Co., Ltd., China), and resuspended in 5% albumin-physiological saline for the following tests.
4. 自体淋巴细胞扩增的倍数: 4. Multiples of autologous lymphocyte expansion:
使用本发明培养后的人淋巴细胞起始数量为 20ml外周静脉血。 培养 前单个核细胞起始数量约为 0.5 - 5 X 107。 培养后, 细胞数目在两周内平 均可增加 323倍(图 1), 三周至四周培养后细胞可扩增 1000倍以上。 The initial number of human lymphocytes cultured using the present invention was 20 ml of peripheral venous blood. The initial number of mononuclear cells before culture is about 0.5 - 5 X 10 7 . After culture, the number of cells is flat within two weeks. Both can be increased by 323 times (Fig. 1), and the cells can be amplified more than 1000 times after three weeks to four weeks of culture.
5. 扩增后自体淋巴细胞的表型: 5. The phenotype of autologous lymphocytes after expansion:
淋巴细胞的表型检测方法如下: The phenotypic detection method of lymphocytes is as follows:
进行标记的细胞每管 106个细胞, 根据所检测的细胞表型需要, 标记 以下不同组合的抗体。 细胞标记采用 FITC-标记的抗 -CD25, PE-CY5-标记 的抗 -CD4, FITC-标记的抗 -CD16, PE-标记的抗 -CD56, PE-CY5-标记的抗- CD8 (Becton-Dickinson, Mountain View, CA, U.S.A.) , PE-标记抗 -CD8 (ebioscience, San Diego, CA, U.S.A.)和 ECD-标记的抗 -CD3 (Beckman Coulter, Fullerton, CA, U.S.A.) o 相应的同型抗体用于背景校正。 表型分析 使用库尔特流式细胞仪 (COULTER EPICS XL), 软件为 COULTER EXP032 ADC (Beckman Coulter, Fullerton, CA, U.S.A.)。 The labeled cells were 10 6 cells per tube, and the antibodies of the different combinations below were labeled according to the required cell phenotype. Cell markers were labeled with FITC-labeled anti-CD25, PE-CY5-labeled anti-CD4, FITC-labeled anti-CD16, PE-labeled anti-CD56, PE-CY5-labeled anti-CD8 (Becton-Dickinson) , Mountain View, CA, USA), PE-labeled anti-CD8 (ebioscience, San Diego, CA, USA) and ECD-labeled anti-CD3 (Beckman Coulter, Fullerton, CA, USA) o corresponding isotype antibody for Background correction. Phenotypic analysis was performed using a Coulter flow cytometer (COULTER EPICS XL) with the software COULTER EXP032 ADC (Beckman Coulter, Fullerton, CA, USA).
本发明所采用的起始细胞为外周血单个核细胞, 其中 T淋巴细胞约 为 40-70%, NK细胞约为 1-5%。 经过培养后, 细胞的主要成分为 T淋巴 细胞和 NK细胞(>95%)。 其中 CD8+ T细胞的比例约为 50-95% (图 2)。 而 CD8+的 T淋巴细胞和 NK细胞是机体发挥抗肿瘤, 抗病毒作用的最主 要淋巴细胞成分。 同时, 本发明方法培养后的细胞 CD4+ T细胞和 CD8+ T细胞的比例明显下降, 有利于淋巴细胞抗肿瘤作用的发挥。 The starting cells used in the present invention are peripheral blood mononuclear cells, wherein the T lymphocytes are about 40-70%, and the NK cells are about 1-5%. After culture, the main components of the cells are T lymphocytes and NK cells (>95%). The proportion of CD8+ T cells is about 50-95% (Fig. 2). CD8+ T lymphocytes and NK cells are the most important lymphocyte components of the body to exert anti-tumor and anti-viral effects. At the same time, the ratio of CD4+ T cells and CD8+ T cells in the cells cultured by the method of the invention is significantly decreased, which is beneficial to the anti-tumor effect of lymphocytes.
6. 用于肿瘤患者的效果: 6. Effect for patients with cancer:
将按照本发明的方法制备的淋巴细胞回输给病人, 细胞的给药量可 根据临床需要而定, 优选为每次细胞量为 5 X 109。 The lymphocytes prepared according to the method of the present invention are returned to the patient, and the amount of the cells administered can be determined according to clinical needs, preferably 5 X 10 9 per cell.
1)提高肿瘤患者体内抗肿瘤细胞因子 Y干扰素(IFN- Y )的水平。 1) Improve the level of anti-tumor cytokine Y interferon (IFN-γ) in tumor patients.
细胞内细胞因子 Y干扰素(IFN- Y )水平的检测方法如下: The detection method of intracellular cytokine Y interferon (IFN-γ) level is as follows:
自接受 EAAL回输的患者采集淋巴细胞。 将 2 X 106/孔细胞铺于 24 孔板, 使用 PMA (10 ng/ml; Sigma-Aldrich Corp., St. Louis, MO, U.S.A.)和 Ionomycin (1 μ g/ml; Sigma-Aldrich Corp., St. Louis, MO, U.S.A.), 在 monensin (10 μ M; Sigma-Aldrich Corp., St. Louis, MO, U.S.A.)存在的条件 下在刺激 6小时。 收获细胞, 用抗表面分子的单克隆抗体如 ECD-CD3和 PE-CY5-CD8室温染色 15 分钟。 用 PBS洗后, 用 Cytoperm (Becton- Dickinson, Mountain View, CA, U.S.A.)禾口含 0.1% saponin (Sigma-Aldrich Corp., St. Louis, MO, U.S.A.)的渗透液处理 20分钟后, 加入 PE-结合的 抗 -IFN- γ抗体(ebioscience, San Diego, CA, U.S.A.)或同型对照, 4°C孵育 30分钟后, 用 PBS洗后, 同上流式细胞仪分析结果。 Lymphocytes were collected from patients receiving EAAL reinfusion. 2 X 10 6 /well cells were plated in 24-well plates using PMA (10 ng/ml; Sigma-Aldrich Corp., St. Louis, MO, USA) and Ionomycin (1 μg/ml; Sigma-Aldrich Corp. , St. Louis, MO, USA), stimulated for 6 hours in the presence of monensin (10 μM; Sigma-Aldrich Corp., St. Louis, MO, USA). Cells were harvested and stained with monoclonal antibodies against surface molecules such as ECD-CD3 and PE-CY5-CD8 for 15 minutes at room temperature. After washing with PBS, treated with a permeate containing Cytoperm (Becton-Dickinson, Mountain View, CA, USA) and 0.1% saponin (Sigma-Aldrich Corp., St. Louis, MO, USA) for 20 minutes, then added to PE. -combined Anti-IFN-γ antibody (ebioscience, San Diego, CA, USA) or isotype control, after incubation at 4 ° C for 30 minutes, washed with PBS, and analyzed by flow cytometry.
在使用 EAAL方法治疗的肿瘤患者体内, 回输后, 其分泌抗肿瘤细 胞因子 DFN- Y的淋巴细胞数目明显增多(图 3, 左)。 同时, 分泌 IFN- Y 的量明显上升(图 3, 右)。 In tumor patients treated with the EAAL method, the number of lymphocytes secreting the anti-tumor cytokine DFN-Y was significantly increased after reinfusion (Fig. 3, left). At the same time, the amount of secreted IFN-γ increased significantly (Fig. 3, right).
IFN- γ除本身具有抑制和杀伤肿瘤的作用外, 对机体免疫系统还具 有重要的调节作用。 它可使免疫系统的功能趋向于 Thl方面平衡, 即发 挥杀伤性功能。 而肿瘤病人体内由于肿瘤分泌的免疫抑制因子的作用, 机体免疫环境多为免疫杀伤抑制性环境, 阻碍了杀伤性淋巴细胞对肿瘤 细胞的杀伤。 因此, EAAL在抗肿瘤治疗的作用机制除直接杀伤或抑制 肿瘤生长外, 可能还包括改善患者体内的免疫抑制性环境, 优化肿瘤杀 伤的免疫环境(见下文 (2))。 In addition to its role in inhibiting and killing tumors, IFN-γ also plays an important role in regulating the body's immune system. It can make the immune system's function tend to balance Thl, which is a killing function. In the tumor patients, due to the action of immunosuppressive factors secreted by tumors, the immune environment of the body is mostly an immunosuppressive environment, which hinders the killing of tumor cells by killer lymphocytes. Therefore, in addition to direct killing or inhibition of tumor growth, the mechanism of action of EAAL in anti-tumor therapy may include improving the immunosuppressive environment in patients and optimizing the immune environment for tumor killing (see (2) below).
2)调节肿瘤患者免疫系统, 明显提高具有抗肿瘤作用的淋巴细胞在 免疫系统中的比例。 2) Regulate the immune system of tumor patients, and significantly increase the proportion of lymphocytes with anti-tumor effects in the immune system.
细胞表型的分析方法同前。 The analysis of cell phenotype is the same as before.
EAAL回输后, 病人外周血淋巴细胞中 CD8+ T细胞的比例明显上升 (图 4), CD4+ T细胞与 CD8+ T细胞的比例明显下降(图 5)。 说明病人的 免疫系统趋向于向免疫杀伤功能的淋巴细胞平衡。 After EAAL reinfusion, the proportion of CD8+ T cells in peripheral blood lymphocytes of patients increased significantly (Fig. 4), and the ratio of CD4+ T cells to CD8+ T cells decreased significantly (Fig. 5). This indicates that the patient's immune system tends to balance lymphocytes to the immune killing function.
3)在部分肿瘤患者中显示出消除肿瘤的作用。 3) It shows the effect of eliminating tumors in some tumor patients.
经过 EAAL回输治疗, 部分患者显示出肿瘤部分消退, 肿瘤稳定, 和生活质量的明显改善的治疗效果(见病例报告及表 1)。 病例报告 1 After EAAL reinfusion treatment, some patients showed partial regression of tumors, stable tumors, and a significant improvement in quality of life (see case report and Table 1). Case report 1
非小细胞肺癌脑转移患者: 该患者在接受 EAAL治疗期间和前三个 月没有接受其它任何治疗, 治疗前后脑 CT显示, 转移灶缩小。 病例报告 2 Patients with non-small cell lung cancer with brain metastases: The patient did not receive any other treatment during the first three months of treatment with EAAL. Brain CT before and after treatment showed a reduction in metastases. Case report 2
复发的乳腺癌, 治疗后(5次回输)肿瘤标志物 CA15.3水平下降(图 Recurrent breast cancer, after treatment (5 times return) tumor markers CA15.3 levels decreased (Figure
6)。 病例报告 3 6). Case report 3
小脑蚓部髓母细胞瘤术后第二次脑脊髓广泛种植复发, 血象一直较 低, 无法正常整套的化疗疗程。 其主管医生采用 EAAL疗法配合化疗, 即每次化疗后 7天, 回输 EAAL, 每周一次, 连续多次。 最后一次回输 后 2周开始, 做下一次化疗。 该患者在 5个月时间内, 经历了 2个化疗 疗程后, 6次 EAAL治疗后, 核磁共振结果显示: 转移灶大部分消失。 After the second epithelial medulloblastoma of the cerebellum, the brain has been replanted extensively, and the blood picture has been low. It is impossible to complete the complete course of chemotherapy. The doctor in charge used EAAL therapy in combination with chemotherapy, ie 7 days after each chemotherapy, EAAL was returned, once a week, several times in succession. Beginning 2 weeks after the last return, do the next chemotherapy. After 5 years of chemotherapeutic treatment and 6 EAAL treatments in 5 months, the NMR results showed that most of the metastases disappeared.
患者在化疗间期的自觉生活质量较第一次复发治疗时明显改善, 治 疗期间不需像第一次复发化疗那样做无菌隔离, 患者在化疗间期一直坚 持爬山锻炼, 没有出现过因贫血导致的心慌、 气短等症状, 无食欲下 降、 体重减轻、 睡眠不良等化疗副反应症状。 病例报告 4 The patient's conscious quality of life during the chemotherapy interval was significantly improved compared with the first relapse treatment. During the treatment period, there was no need for aseptic isolation like the first relapse chemotherapy. The patient insisted on climbing the mountain during the chemotherapy interval, and there was no abnormality due to anemia. Symptoms such as palpitation, shortness of breath, and other symptoms of chemotherapy side effects such as decreased appetite, weight loss, and poor sleep. Case report 4
结肠癌双肺转移患者, 女, 65岁, 化疗 +EAAL回输治疗 Colon cancer with double lung metastases, female, 65 years old, chemotherapy + EAAL reinfusion treatment
病人食睡正常, 精神体力良好, 咳嗽消失, 复査显示肺部病灶有所 缩小, 多个淋巴结转移有所好转。 The patient had normal food and sleep, good mental strength, and cough disappeared. The review showed that the lung lesions had shrunk and multiple lymph node metastases had improved.
表 1.部分患者经 EAAL回输治疗后的临床效果。 Table 1. Clinical effects of some patients after EAAL reinfusion treatment.
患者编号 肿瘤类型 其他治疗 临床效果 Patient Number Tumor Type Other Treatment Clinical Effects
0002 非小细胞肺癌伴胸膜转 EAAL治疗前 4个月 1. 5次细胞回输结束 12天后脑部病灶缩小, 肺部病灶无进展 0002 Non-small cell lung cancer with pleural effusion 4 months before EAAL treatment 1. After 5 times of cell reinfusion, brain lesions shrink after 12 days, no progress in lung lesions
移无法手术, 后伴脑转 曾进行放疗 2. 生活质量改善: 细胞回输后, 曾持续一年的每日下午低热消 移 失, 食欲和睡眠改善。 Immobilization, post-brain transfer, and radiotherapy 2. Improvement in quality of life: After the cell was returned, it continued for one year of daily low heat loss, appetite and sleep improvement.
0005 非小细胞肺癌伴淋巴结 手术, 化疗 1. 手术后 6个月疾病无进展 0005 Non-small cell lung cancer with lymph nodes surgery, chemotherapy 1. No progress after 6 months of surgery
浸润 2. 生活质量改善: 化疗造成的不良反应得到缓解 Infiltration 2. Quality of life improvement: adverse reactions caused by chemotherapy are alleviated
0015 非小细胞肺癌伴骨转移 胸腺肽治疗 1. 6次回输后乙肝表面抗原降低,疼痛缓解, 胸水消失 0015 Non-small cell lung cancer with bone metastasis Thymosin treatment 1. Hepatitis B surface antigen decreased after 6 times of reinfusion, pain relief, pleural effusion disappeared
分子靶向疗法 2. 治疗后 4个月咳嗽渐加重但胸片无显示明显病灶 Molecular targeted therapy 2. After 4 months of treatment, the cough gradually worsened but the chest radiograph showed no obvious lesions.
0017 非小细胞肺癌 无 1. 前 2次回输后胸水显著减少 0017 Non-small cell lung cancer None 1. Significant reduction in pleural effusion after the first 2 reinfusions
2. 第 3、 4次回输后胸水无变化, 疾病无进展 2. There is no change in pleural effusion after the third and fourth reincarnation, no progress in disease
0010 贲门癌伴肝转移, 腹腔 治疗前 1个月进行化 1. 前 2次细胞回输后腹水显著减少, 肝及腹膜转移灶部分缩小 转移 疗, 后无其他治疗 0010 Cardiac cancer with liver metastasis, 1 month before treatment of abdominal cavity 1. After the first 2 times of cell reinfusion, the ascites was significantly reduced, and the liver and peritoneal metastases were partially reduced. After treatment, there was no other treatment.
0032 结肠癌伴腹腔转移 中药治疗 1.生活质量改善 0032 Colon cancer with abdominal cavity transfer Chinese medicine treatment 1. Quality of life improvement
2.体重增加 2. Weight gain
0031 乳腺癌伴肝、 骨转移 治疗前化疗刚结束 1.大肿瘤灶缩小, 小肿瘤灶无变化 0031 Breast cancer with liver and bone metastasis Just before the treatment of chemotherapy 1. Large tumors shrink, small tumors have no change
0027 髓母细胞瘤术后, 多病 化疗 1. 两次化疗配合六次高效 CIK治疗后脑部病灶基本完全.消失 0027 Postoperative medulloblastoma, multiple disease chemotherapy 1. Two chemotherapy combined with six high-efficiency CIK treatments, the brain lesions are completely complete.
灶第二次复发 The second recurrence of the stove
0029 肺癌术后累及脏层胸 口服分子靶向药物 1. 双肺转移结节明显好转 膜, 双肺转移后化疗无 2. 左侧胸膜增厚结节好转 0029 Lung cancer postoperative involvement of visceral chest Oral molecular targeted drugs 1. Double lung metastasis nodules improved significantly, no chemotherapy after double lung metastasis 2. Left pleural thickening nodules improved
法控制 3. 双侧胸水吸收 Method control 3. bilateral pleural effusion
0035 胃癌术后复发, 腹膜转 无 1. 进食后哽咽感消失, 食欲有所增加 0035 Postoperative recurrence of gastric cancer, peritoneal rotation None 1. After eating, the pharyngeal sensation disappears and the appetite increases.
移 2. CT复查病情无进展 Move 2. CT review no progress
0039 右肺癌术后, 肝转移 无 1. 生活质量改善 0039 Right lung cancer postoperative, liver metastasis None 1. Quality of life improved
2. 体重增加 2. Weight gain
0041 右肺癌, 纵隔淋巴结肿 无 1. 生活质量改善 0041 Right lung cancer, mediastinal lymphadenopathy None 1. Quality of life improvement
大、 心包积液 2. 复査显示肿瘤标志物下降 Large, pericardial effusion 2. Reexamination showed a decline in tumor markers
0046 结肠癌双肺转移 口服化疗药, 中药 1. 咳嗽消失 0046 Colon cancer double lung metastasis Oral chemotherapy, Chinese medicine 1. Cough disappears
2. 肺部病灶缩小, 多个淋巴结转移有所好转 2. The lung lesions shrink, and multiple lymph node metastases have improved.
Claims
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN200710102854.0 | 2007-05-09 | ||
| CN2007101028540A CN101302491B (en) | 2007-05-09 | 2007-05-09 | Highly effective method for amplifying activated lymphocyte and cultivation system |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2008138214A1 true WO2008138214A1 (en) | 2008-11-20 |
Family
ID=40001680
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2008/000887 Ceased WO2008138214A1 (en) | 2007-05-09 | 2008-04-30 | A method for activation and expansion of lymphocytes with high efficiency and the culture sysytem thereof |
Country Status (2)
| Country | Link |
|---|---|
| CN (1) | CN101302491B (en) |
| WO (1) | WO2008138214A1 (en) |
Cited By (7)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2013094988A1 (en) | 2011-12-22 | 2013-06-27 | Mogam Biotechnology Research Institute | Method for producing natural killer cells, natural killer cells produced thereby, and composition for treating cancers and infectious diseases containing the same |
| US20160289624A1 (en) * | 2013-12-18 | 2016-10-06 | Toyo Seikan Group Holdings, Ltd. | Culture container, method for culturing lymphocytes, culture-container production method, and solid-phasing apparatus |
| CN109593713A (en) * | 2018-12-29 | 2019-04-09 | 广州和能生物科技有限公司 | A kind of cultural method of autologous peripheral blood lymphocyte |
| CN115508557A (en) * | 2022-06-08 | 2022-12-23 | 河南道特尔医药科技股份有限公司 | Method for testing antibody secretion effect of positive B cell of hepatitis B antibody on hepatitis B vaccine |
| US11766456B2 (en) | 2014-11-26 | 2023-09-26 | GC Cell Corporation | Method for culturing natural killer cells using T cells |
| US12203065B2 (en) | 2017-05-26 | 2025-01-21 | GC Cell Corporation | Method for culturing natural killer cell, using transformed T cell |
| US12398372B2 (en) | 2018-11-14 | 2025-08-26 | GC Cell Corporation | Method for culturing cord blood-derived natural killer cells using transformed T-cells |
Families Citing this family (10)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN102676453A (en) * | 2011-03-17 | 2012-09-19 | 中国医学科学院肿瘤研究所 | Method for culturing natural killer (NK) and/or natural killer T (NKT) cells |
| JP6405690B2 (en) * | 2014-05-09 | 2018-10-17 | 東洋製罐グループホールディングス株式会社 | Multi-chamber culture container and cell culture method |
| CN104673750B (en) * | 2015-02-13 | 2018-04-27 | 杭州易文赛生物技术有限公司 | A kind of method of natural killer cells amplification and a kind of culture media composition |
| CN105969731B (en) * | 2016-07-29 | 2019-10-22 | 青岛市中心医院 | A method for preparing a large amount of TIL cells with high killing activity by using malignant pleural effusion |
| CN106854644A (en) * | 2017-03-08 | 2017-06-16 | 广州可伴咨询中心(普通合伙) | A kind of in vitro culture kit of CMV-CTL cells |
| CN108949684B (en) * | 2018-07-02 | 2020-08-18 | 西安交通大学 | CIK cell and preparation method and application thereof |
| US20250064931A1 (en) * | 2021-12-28 | 2025-02-27 | Immunotech Applied Science Limited | Activated lymphocyte expansion method having stable and controllable quality, and anti-tumor use thereof |
| WO2023125704A1 (en) * | 2021-12-28 | 2023-07-06 | 北京永泰生物制品有限公司 | Activated lymphocyte expansion method having stable and controllable quality, and use thereof for prevention and control of neurological disease |
| CN114984048A (en) * | 2022-06-16 | 2022-09-02 | 浙江百越生物技术有限公司 | Cell therapy for preventing and treating senile tarnish |
| CN115261321B (en) * | 2022-09-27 | 2022-12-27 | 北京大学口腔医学院 | Method for enhancing anti-tumor function of T lymphocyte and application |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1444648A (en) * | 2000-06-06 | 2003-09-24 | 麒麟麦酒株式会社 | Methods of expanding natural killer T cells |
| CN1503678A (en) * | 2001-04-26 | 2004-06-09 | 3 | Use of activated peripheral blood mononuclear cells for the treatment of cancer |
| CN1703506A (en) * | 2002-10-04 | 2005-11-30 | 埃比奥吉恩药物股份公司 | Procedure for the large-scale t-lymphocytes culture in a homogeneous system |
Family Cites Families (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1116549A (en) * | 1994-08-09 | 1996-02-14 | 中国医学科学院肿瘤研究所 | High affinity tumor killer cell (T-AK cell) preparation |
| JP2003530826A (en) * | 1999-11-17 | 2003-10-21 | ユニバーシティー オブ ロチェスター | Human ex vivo immune system |
-
2007
- 2007-05-09 CN CN2007101028540A patent/CN101302491B/en active Active
-
2008
- 2008-04-30 WO PCT/CN2008/000887 patent/WO2008138214A1/en not_active Ceased
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1444648A (en) * | 2000-06-06 | 2003-09-24 | 麒麟麦酒株式会社 | Methods of expanding natural killer T cells |
| CN1503678A (en) * | 2001-04-26 | 2004-06-09 | 3 | Use of activated peripheral blood mononuclear cells for the treatment of cancer |
| CN1703506A (en) * | 2002-10-04 | 2005-11-30 | 埃比奥吉恩药物股份公司 | Procedure for the large-scale t-lymphocytes culture in a homogeneous system |
Non-Patent Citations (2)
| Title |
|---|
| HIROSHI AZUMA ET AL.: "Functional evaluation of ex vivo expanded cord blood lymphocytes: Possible use for adaptive cellular immunotherapy", EXPERIMENTAL HEMATOLOGY, vol. 30, 31 December 2002 (2002-12-31), pages 346 - 351, XP002411884 * |
| ROBINSON K.L. ET AL.: "Ex vivo expansion, maturation, and activation of umbilical cord blood-derived T lymphocytes with IL-2, IL-12, anti-CD3, and IL-7: Potential for adoptive cellular immunotherapy post-umbilical cord blood transplantation", EXPERIMENTAL HEMATOLOGY, vol. 30, 2002, pages 245 - 251, XP002411885 * |
Cited By (11)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2013094988A1 (en) | 2011-12-22 | 2013-06-27 | Mogam Biotechnology Research Institute | Method for producing natural killer cells, natural killer cells produced thereby, and composition for treating cancers and infectious diseases containing the same |
| EP2794859A4 (en) * | 2011-12-22 | 2015-05-27 | Mogam Biotechnology Inst | METHOD FOR PRODUCING NATURAL KILLER CELLS, NATURAL KILLER CELLS PRODUCED THEREFOR, AND COMPOSITION FOR TREATING CANCERS AND INFECTIOUS DISEASES CONTAINING THE SAME |
| US20160289624A1 (en) * | 2013-12-18 | 2016-10-06 | Toyo Seikan Group Holdings, Ltd. | Culture container, method for culturing lymphocytes, culture-container production method, and solid-phasing apparatus |
| US11884907B2 (en) * | 2013-12-18 | 2024-01-30 | Toyo Seikan Group Holdings, Ltd. | Culture container, method for culturing lymphocytes, culture-container production method, and solid-phasing apparatus |
| US12264307B2 (en) | 2013-12-18 | 2025-04-01 | Toyo Seikan Group Holdings, Ltd. | Culture container, method for culturing lymphocytes, culture-container production method, and solid-phasing apparatus |
| US11766456B2 (en) | 2014-11-26 | 2023-09-26 | GC Cell Corporation | Method for culturing natural killer cells using T cells |
| US12203065B2 (en) | 2017-05-26 | 2025-01-21 | GC Cell Corporation | Method for culturing natural killer cell, using transformed T cell |
| US12398372B2 (en) | 2018-11-14 | 2025-08-26 | GC Cell Corporation | Method for culturing cord blood-derived natural killer cells using transformed T-cells |
| CN109593713A (en) * | 2018-12-29 | 2019-04-09 | 广州和能生物科技有限公司 | A kind of cultural method of autologous peripheral blood lymphocyte |
| CN115508557A (en) * | 2022-06-08 | 2022-12-23 | 河南道特尔医药科技股份有限公司 | Method for testing antibody secretion effect of positive B cell of hepatitis B antibody on hepatitis B vaccine |
| CN115508557B (en) * | 2022-06-08 | 2025-05-16 | 河南道特尔医药科技股份有限公司 | A method for testing the antibody secretion effect of hepatitis B antibody-positive B cells on hepatitis B vaccine |
Also Published As
| Publication number | Publication date |
|---|---|
| CN101302491A (en) | 2008-11-12 |
| CN101302491B (en) | 2011-09-14 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2008138214A1 (en) | A method for activation and expansion of lymphocytes with high efficiency and the culture sysytem thereof | |
| CN108699523B (en) | NK cell culture medium addition kit and NK cell culture method using same | |
| JP5358683B2 (en) | Method of growing natural killer cells | |
| JP3056230B2 (en) | Peripheral blood lymphocyte cell proliferation stimulation method | |
| CN108220235A (en) | A kind of Activated in Vitro expands people's natural kill(NK)The method of cell and its special cultivating system | |
| WO2015014291A1 (en) | Lymph cell amplification and activation method via serum-free cultivation | |
| JP3619853B2 (en) | Method of growing natural killer cells | |
| CN105886469B (en) | CIK cell and its cultural method and application | |
| JP2017061558A (en) | Cells expressing Th1 properties and cytolytic properties | |
| CN106589133A (en) | Preparation and applications of liver-cancer-specific CTL cells induced by new enhanced antigen combined polypeptide | |
| CN113528434A (en) | A universal high-efficiency in vitro expansion method for clinical multiple reinfusion of allogeneic DNT cells | |
| CN113980901B (en) | Method for preparing high-purity mature human dendritic cells and application | |
| CN116179486A (en) | Preparation method of tumor-infiltrating lymphocytes | |
| CN114231486A (en) | Generation of HPV antigen-specific T cells from naive T cell populations | |
| CN107849537A (en) | Optimal activated dendritic cells that induce improved or increased anti-tumor immune responses | |
| CN110747167B (en) | Preparation method and application of hemizygous BAK cell | |
| TW200404002A (en) | Treatment method achieved by using the lymphocytes derived from HLA matching donor-originating activated lymphocytes, formula having the lymphocytes as a main constituent thereof; method and preparation kit for manufacturing the formula | |
| JP2025072455A (en) | Method for producing highly pure and highly efficient naturally killed cells and its use | |
| US20100092499A1 (en) | Alpha Thymosin Peptides as Cancer Vaccine Adjuvants | |
| CN1990044A (en) | Preparation and use of human body immunocompetent cell DCCIK anti-tumor cell preparation | |
| CN107708727A (en) | It is a kind of to be used to treat tumor vaccine of liver cancer and preparation method thereof | |
| CN110205293A (en) | A kind of preparation method and application of the NK immunocyte of reinforced efficient treatment lung cancer | |
| CN103602634A (en) | Preparation method of DCs, and application of DCs in preparing anti-tumor cell preparation | |
| CN113293130A (en) | Culture method of tumor specific T cells | |
| JP4143772B2 (en) | Postoperative recurrence preventive agent for intravenous administration of glioblastoma and method for producing peripheral blood activated lymphocytes collected from glioblastoma patients for preventing postoperative recurrence of glioblastoma |
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: 08748446 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 08748446 Country of ref document: EP Kind code of ref document: A1 |