CN111909899A - Method for enriching T cells and application of method in adoptive T cell therapy - Google Patents

Method for enriching T cells and application of method in adoptive T cell therapy Download PDF

Info

Publication number
CN111909899A
CN111909899A CN201910375997.1A CN201910375997A CN111909899A CN 111909899 A CN111909899 A CN 111909899A CN 201910375997 A CN201910375997 A CN 201910375997A CN 111909899 A CN111909899 A CN 111909899A
Authority
CN
China
Prior art keywords
cells
cell
pbmcs
car
magnetic beads
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201910375997.1A
Other languages
Chinese (zh)
Inventor
鲁薪安
何霆
齐菲菲
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beijing Yimiao Shenzhou Pharmaceutical Technology Co ltd
Original Assignee
Beijing Yimiao Shenzhou Pharmaceutical Technology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beijing Yimiao Shenzhou Pharmaceutical Technology Co ltd filed Critical Beijing Yimiao Shenzhou Pharmaceutical Technology Co ltd
Priority to CN201910375997.1A priority Critical patent/CN111909899A/en
Publication of CN111909899A publication Critical patent/CN111909899A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15021Viruses as such, e.g. new isolates, mutants or their genomic sequences

Landscapes

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

Abstract

The invention relates to the technical field of adoptive T cell immunotherapy, in particular to a method for enriching T cells and application thereof in adoptive T cell therapy. The enrichment method comprises the steps of culturing Peripheral Blood Mononuclear Cells (PBMCs) for 1-4 hours, removing adherent cells, and then separating T cells from the rest cells by using magnetic beads. The method can effectively improve the purity of the T cells, and has high T cell yield and simple operation. The invention also improves the expression rate of the Chimeric Antigen Receptor (CAR) and reduces the operation risk by changing a virus infection method.

Description

Method for enriching T cells and application of method in adoptive T cell therapy
Technical Field
The invention relates to the technical field of adoptive T cell therapy, in particular to a method for enriching T cells and application of the method in preparing T cells for adoptive cell therapy.
Background
In recent years, there has been widespread interest and use of tumor immunotherapy, which controls and eliminates malignant cells by activating the body's anti-tumor immune response (Sanmamed MF, et al. cell.2019,176: 677). Cancer cells are transformed from normal cells by genetic mutations or epigenetic changes, which express many abnormal tumor antigens. Normally, the innate immune system of the human body specifically recognizes and kills cancer cells by recognizing tumor antigens. However, cancer tissues adopt various mechanisms to construct immunosuppressive microenvironments that allow immune cells such as T and B lymphocytes to develop immune tolerance against cancer cells, leading to malignant progression (Kalathil SG, et al cancer immunological impronother.2016, 65(7): 813). Compared with the traditional therapy, the tumor immunotherapy has the remarkable advantages of good curative effect and small toxic and side effect by breaking immune tolerance, restarting and maintaining tumor-immune circulation.
Adoptive T cell therapy is an emerging tumor immunotherapy that utilizes in vitro enrichment and engineered modification of human T cells to specifically target and kill cancer cells in patients (Yang JC, et al. adv immunol.2016,130: 279). The adoptive T cell therapy currently available includes tumor infiltrating lymphocytes, chimeric antigen receptor T cells (CAR-T), T cell receptor chimeric T cells (TCR-T), and the like. Various techniques have been developed at home and abroad to enrich naturally occurring T cells capable of targeting tumor antigens, or to specifically target known tumor antigens by genetically modifying T cells. Clinical studies have demonstrated that adoptive T cell therapy significantly inhibits cancer progression, prolongs patient survival, and in particular the emergence of CAR-T technology, has led to a milestone development in human control of cancer (June CH, et al science.2018,359: 1361). The application of this technology in tumor therapy started in 1989, and has achieved a response rate of 60-95% in hematological malignancies such as relapsed refractory leukemia, lymphoma, multiple myeloma (Park JH, et al. N Engl J Med 2018,378: 449; Neelapu SS, et al. N Engl J Med 2017,377: 2531; Cohen AD, et al. J Clin invest.2019,130: pii:126397), and also has shown potential therapeutic effects in some malignant solid tumors such as glioma, liver Cancer, melanoma (Migliorini D, et al. Clin Cancer Res.2018,24: 535; Gao H, et al. Clin Cancer Res.2014,20: 6418; Forsberg EMV, et al. Cancer Res.2019,79: 899).
The T cells currently used in CAR-T technology are mainly derived from the patient himself and are greatly affected by the condition of the patient. T cells were isolated by three methods: firstly, magnetic beads are directly separated from PBMCs; collecting cells from the PBMCs by a method of multiple adherence, and adding magnetic beads for activation; collecting cells from PBMCs by a method of multiple adherence, adding magnetic beads for activation, and separating T cells by using the magnetic beads after activation. The T cells separated by the first two methods have low purity, the activation efficiency, CAR expression rate and cell proliferation efficiency of the T cells are influenced, the sorting in the third method influences the activity of the cells after activation, the T cell yield is reduced, and the controllability on the number of the infectable T cells is low. In addition, the T cells separated by the prior art have the problems of low expression rate of target proteins and the like in the subsequent virus infection process. Therefore, methods for separating and purifying T cells and infecting viruses need to be improved, which is important for improving the success rate of clinical application of adoptive T cell therapy.
In view of the above, the present invention is particularly proposed.
Disclosure of Invention
The invention relates to a method for enriching T cells, which comprises the steps of culturing PBMCs for 1-4 hours, removing adherent cells, and then separating the T cells from the rest cells by using magnetic beads.
T cells isolated from PBMCs have low purity and contain a part of adherent cells such as macrophages and dendritic cells, B cells and the like, which results in low T cell activation efficiency, low virus infection efficiency and unclear target cell components. The T cell enrichment method provided by the invention can effectively improve the purity of the T cells, and has high T cell yield and simple operation.
According to one aspect of the invention, the invention also relates to the use of a method as described above for the preparation of a medicament for T cell immunotherapy;
the T cell immunotherapy drug comprises one or more of the following cells: tumor infiltrating lymphocytes, cytotoxic T cells, chimeric antigen receptor T cells (CAR-T), T cell receptor chimeric T cells, natural killer T cells, and peripheral blood lymphocytes.
According to one aspect of the invention, the invention also relates to a method of making a CAR-T cell comprising:
i) enriching T cells using the methods described above;
ii) inoculating the virus carrying the CAR expression fragment 24-48 hours after T cell activation; and
iii) culturing the T cells.
According to the invention, the CAR expression rate is improved and the operation risk is reduced by changing a virus infection method.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below, and it is obvious that the drawings in the following description are some embodiments of the present invention, and other drawings can be obtained by those skilled in the art without creative efforts.
FIG. 1 is a graph showing T cell proliferation isolated from PBMCs from different sources in one embodiment of the present invention;
FIG. 2 is a flow chart of monocyte depletion after PBMCs are attached in one embodiment of the invention; BNK: before adhering to the wall; 1-BNK: after adhering to the wall;
FIG. 3 is a graph of T cell sorting using different ratios of beads to T cells in one embodiment of the invention;
FIG. 4 is a graph of the effect of infection cofactors on CAR expression rate in one embodiment of the invention;
FIG. 5 is a graph showing the effect of different IL-2 concentrations on cell proliferation in one embodiment of the present invention;
FIG. 6 is a culture expansion of three patient-derived CAR-T cells in one embodiment of the invention.
Detailed Description
According to one aspect of the present invention, the present invention relates to a method for enriching T cells, which comprises culturing PBMCs for 1 to 4 hours, removing adherent cells, and separating T cells from the remaining cells using magnetic beads.
Lymphocytes are suspension cells or minimally adherent, and adherent treatment can remove mixed cells such as macrophages in the PBMCs.
The invention only carries out one-time treatment adherent culture, and the loss of T cells is less.
PBMCs refer to cells with a single nucleus in peripheral blood, including lymphocytes and monocytes.
In the present invention, unless otherwise specified, the cell culture conditions are all 30 ℃ to 45 ℃ and 1% to 10% CO2Preferably 36-38 ℃ and 4-6% CO2
Specific subsets of the sorted T cells may be selected for example from CD28+、CD4+、CD8+、CD45RA+And CD45RO+Cells of any positive combination in T cells; in some embodiments, the T cell selected is CD3+CD28+T cells.
The above subpopulations of T cells may be further isolated by positive or negative selection techniques. For example CD3+CD28+T cells can be positively selected using CD3/CD28 conjugated magnetic beads (e.g., Dynabeads CD3/CD 28). In some embodiments, the magnetic beads conjugated with CD3/CD28 antibody are mixed with the remaining cells at a magnetic bead: t cells are incubated at a ratio of 1.5:1, 2:1, or 2.5:1, for example, and then sorted using the magnetic beads.
Wherein (1-3): 1 is the ratio of the number of magnetic beads to the number of T cells. The number of T cells and the estimation of the number of T cells by conventional methods such as morphological observation of the remaining cells.
In some embodiments, the PBMCs are cultured at a seeding density of (0.5-1.5). times.108cells/30 mL; preferably 1X 108cells/30mL。
In some embodiments, the culture area of cells available in the culture flask for said adherent culture is 175cm2In terms of volume, the addition amount of the culture medium used for adherent culture is 25-35 mL, and 30mL is preferably selected.
In some embodiments, the sorting is performed at a cell density of (0.15-0.6) x 106cells/mL, preferably (0.25-0.5). times.106cells/mL。
In some embodiments, the paramagnetic microparticles used in the present invention are of a size sufficient to be phagocytized by phagocytic monocytes, which are subsequently removed by magnetic separation. In certain embodiments, the paramagnetic microparticles are commercially available beads, for example, those manufactured by Dynal AS under the trade name DynabeadsTMThe beads of (a). Dynabeads, as exemplified hereinTMIs M-280, M-450 and M-500. In one aspect, paramagnetic microparticles coated with "irrelevant" proteins (e.g., serum proteins or antibodies) remove other non-specific cells. Unrelated proteins and antibodies include those that do not specifically target the T cells to be expanded or fragments thereof. In certain embodiments, the unrelated beads include beads coated with sheep anti-mouse antibodies, goat anti-mouse antibodies, and human serum albumin.
In one aspect of the invention, enrichment of T cell populations by negative selection is accomplished with a combination of antibodies directed to cell surface unique markers of the negative selection. The preferred method is cell sorting and/or selection by negative magnetic immunoadhesion or flow cytometry using a mixture of monoclonal antibodies directed against cell surface markers on negatively selected cells. For example, enrichment of CD4 by negative selection+The cell, monoclonal antibody cocktail generally includes antibodies against CD14, CD20, CD11b, CD16, HLA-DR, and CD 8.
T cells can be obtained from xenogeneic sources, e.g., mouse, rat, non-human primate, and pig.
In some embodiments, the PBMCs are from a subject in need of a T cell therapy.
In some embodiments, the PBMCs are from a single blood draw, peripheral blood whole blood, or peripheral blood stem cells.
At present, T cells used in the CAR-T culture process are all derived from patients, some patients are not suitable for single blood collection according to the condition of the patients, and some patients have low T cell activity and slow proliferation speed and cannot be cultured and expanded to the dose required by patient feedback, so that the patients cannot receive CAR-T treatment finally.
Preferably, the cells are isolated from the circulating blood of the individual by apheresis or leukapheresis. Apheresis typically contains lymphocytes (T cells, monocytes, granulocytes, B cells), other nucleated leukocytes, erythrocytes, and platelets. In one embodiment, cells collected by apheresis or leukapheresis may be washed to remove plasma components and placed in an appropriate buffer or culture medium for subsequent processing steps. In one embodiment of the invention, the cells are washed with phosphate-buffered saline (PBS). In further embodiments, the wash solution may lack calcium, may lack magnesium, may lack all divalent ions, or lack many divalent ions. As those of ordinary skill in the art will readily appreciate, the washing step may be accomplished by methods well known in the art, such as by using semi-automated "extreme sedimentation" centrifugation according to the manufacturer's recommendations. After washing, the cells can be resuspended in various biocompatible buffers, e.g., Ca-free2+/Mg2+The PBS (1). In addition, unwanted components of the apheresis sample can be removed and the cells resuspended in culture medium.
In some embodiments, the PBMCs are freshly isolated cells.
In some embodiments, the PBMCs are cells that are thawed after cryopreservation.
According to one aspect of the invention, the invention also relates to the use of a method as described above for the preparation of a medicament for T cell immunotherapy;
the T cell immunotherapy drug comprises one or more of the following cells: tumor infiltrating lymphocytes, cytotoxic T cells, chimeric antigen receptor T cells, T cell receptor chimeric T cells, natural killer T cells and peripheral blood lymphocytes.
According to one aspect of the invention, the invention also relates to a method of making a CAR-T cell comprising:
i) enriching T cells using the methods described above;
ii) inoculating the virus carrying the CAR expression fragment 24-48 hours after T cell activation; and
iii) culturing the T cells.
T cell activation can be performed by co-incubation with magnetic beads conjugated with CD3/CD28 antibodies as described above, or by the addition of certain well-known cytokine/cytokine antibodies that promote activation.
The term "inoculation" as used herein refers to the addition of virus to a host cell to initiate culture; or adding cells to the initial medium to start the culture.
Preferably, in step ii), the virus carrying the CAR-expressing fragment is inoculated 36-48 hours after T cell activation; alternatively, 40 hours, 43 hours, 44 hours or 46 hours, most preferably 48 hours, may be used.
In some embodiments, the virus carrying the CAR expression fragment is a retrovirus.
In some embodiments, the virus carrying the CAR expression fragment is a lentivirus.
In some embodiments, the virus carrying the CAR expression fragment is inoculated only once.
By "vaccinate once" is meant that the virus is added only once within 24 hours.
In some embodiments, no infection co-factor is added during the process of making the CAR-T cells.
The infection auxiliary factor is selected from, for example, Vectofusin-1 and Polybrene.
In some embodiments, in step iii), the cells are cultured using serum-free, antibiotic-free medium.
At present, CAR-T culture mostly adopts a culture medium containing serum and antibiotics, the serum source is AB serum or autologous serum, and great potential safety hazards exist. The serum-free and antibiotic-free culture medium used in the CAR-T culture process reduces the influence of serum and antibiotics on cells in the culture process and ensures that serum-free factors and antibiotics are not contained in the reinfusion preparation. The safety of the product is increased.
In some embodiments, the serum-free medium that can be used is selected from the group consisting of X-VIVO15 medium from Lonza.
In some embodiments, the medium contains 100-600 IU/mL Interleukin-2 (Interleukin 2, IL-2);
in some embodiments, the medium contains 500IU/mL IL-2.
IL-2 is also known as T Cell Growth Factor (TCGF) because it enables long-term, sustained T cell proliferation in vitro. Quiescent T cells do not express IL-2R and therefore cannot serve as target cells for IL-2; the surface of T cells activated by mitogen or other stimuli can express IL-2R and respond to IL-2 stimulation. IL-2 can enhance the killing activity of T cells, and can induce the generation of cytotoxic T cells (Tc or CTL) together with IL-4, IL-5 and IL-6 in vitro, so that the activity of the cytotoxic T cells is greatly enhanced; the T cells induced by IL-2 can generate obvious anti-tumor effect after being returned into the body of a patient, but the T cells are not easy to survive in the body, and if a small amount of IL-2 is simultaneously input, the survival time of the T cells in the body can be obviously prolonged, and the anti-tumor effect of the T cells can be enhanced.
In some embodiments, in step iii), the seeding density of T cells is 0.5-0.8 × 106cells/mL;
In some embodiments, in step iii), the seeding density of T cells is 0.6-0.7 × 106cells/mL。
In some embodiments, in step iii), the medium is changed every 12-36 hours (preferably 24 hours), and the ratio of the new medium to the old medium is (5-1): 1, or 4:1, 3:1 or 2: 1.
In some embodiments, in step iii), the incubation time is 7 to 8 days in total.
Embodiments of the present invention will be described in detail below with reference to examples, but it will be understood by those skilled in the art that the following examples are only illustrative of the present invention and should not be construed as limiting the scope of the present invention. The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
EXAMPLE 1 isolation of autologous PBMCs from patients of different origins
The T cells used in the autologous CAR-T culture process are mainly derived from the patient, but in some cases, due to the condition of the patient or the condition of the patient, the PBMCs have low T cell content or insufficient T cell activity, which increases the difficulty of T cell separation and proliferation in the CAR-T production process. The CAR-T production can be carried out on the patients by using peripheral blood stem cells, PBMCs stored in the healthy state of the patients or high-activity PBMCs stored in the early onset stage of the patients, so that the culture failure caused by low activity of T cells can be effectively avoided.
Preparation of PBMCs from different sources
Scheme one is to separate PBMCs from autologous blood sample by using Ficoll separating medium.
The experimental procedure was as follows:
1. blood samples were collected as blood samples: the saline solution was diluted 1:4 by volume.
2. The diluted blood samples were separated using Ficoll medium (800g/20 min/liter 5 down 4).
3. The buffy coat layer was collected into a 50mL centrifuge tube and subjected to trim centrifugation (500g/6 min/L8 down 8).
4. The supernatant was discarded and washed twice with physiological saline (400g/6 min/L8, 8).
Scheme two PBMC is obtained by separating autologous peripheral blood with Ficoll separating medium.
The experimental procedure was as follows:
1. peripheral blood samples were as per blood sample: the saline solution is diluted in a volume ratio of 1: 1.
2. The diluted blood samples were separated using Ficoll medium (800g/20 min/liter 5 down 4).
3. The buffy coat layer was collected into a 50mL centrifuge tube and subjected to trim centrifugation (500g/6 min/L8 down 8).
4. The supernatant was discarded and washed twice with physiological saline (400g/6 min/L8, 8).
And (3) recovering the PBMCs frozen in the third scheme in a water bath, and washing out frozen stock solution to obtain the PBMCs.
The experimental procedure was as follows:
1. taking out the frozen PBMCs, rapidly resuscitating in water bath at 37 ℃, transferring into a 50mL centrifuge tube, balancing and centrifuging (400g/5 min/L8-step down 8)
2. The supernatant was discarded, and PBMCs were washed twice with physiological saline (400g/5 min/liter 8 down)
And recovering the peripheral blood stem cells stored in the fourth scheme in a water bath to obtain the low-purity PBMCs.
The experimental procedure was as follows:
1. the stored cells are taken out and quickly recovered in a water bath at 37 ℃, transferred into a 50mL centrifuge tube, and subjected to ingredient centrifugation (400g/5 min/L8-step-down 8)
2. The supernatant was discarded, and PBMCs were washed twice with physiological saline (400g/5 min/liter 8 down)
T cells isolated using PBMCs from different sources proliferated well during CAR-T culture (FIG. 1). T cells sorted from the PBMCs from four sources can be cultured for CAR-T.
Example 2 obtaining autologous T cells from a patient
In the T cell sorting process, the high-purity T cells obtained by separation have promotion effects on improving the activation efficiency of the T cells, the proliferation multiple of the T cells and the CAR expression rate. However, in the CAR-T production process, PBMCs contain a large number of monocytes due to the patient himself, which results in a low T cell content, adding difficulties to the isolation of T cells to a high purity.
In the scheme, PBMCs are treated in an adherence way, monocytes in the PBMCs are removed, suspension cells are collected, and Dynabeads CD3/CD28 is added to sort T cells.
T cell isolation step:
1. the isolated PBMCs were dispensed in 30 mL/bottle (1X 10 total)8cells) were spread in a T175 flask and placed in a carbon dioxide incubator (37 ℃ C., 5% CO)2) And (5) culturing.
After 2.2 hours, the cells were removed, aspirated and centrifuged (400g/6 min/l 8 down 8).
3. After resuspension, the pellet was resuspended as beads: t-3: 1, 2:1, 1.5:1, 1:1 Dynabeads CD3/CD28 was added and incubated at room temperature for 20 min.
4. After incubation, the cell suspension was diluted to 0.5X 106T cells/mL or 0.25X 106Sorting was performed in T cells/mL.
5. And (4) spreading the sorted T cells into a culture flask for culture.
After PBMCs are attached to the wall, a monocyte removal flow chart is shown in figure 2, and as can be seen from the chart, the monocyte removal efficiency after the PBMCs are attached to the wall is high, and the proportion of lymphocytes is increased. PBMCs adherent treatment, monocyte removal efficiency and lymphocyte yield are shown in Table 1.
Sorting of different beads versus T cells is shown in FIG. 3. As can be seen from the figure, the ratio pairs of 3:1, 2:1, 1.5:1, 1:1 are all suitable for sorting T cells.
TABLE 1
Figure BDA0002051682030000111
Figure BDA0002051682030000121
Wherein, DP073, DP074, DP079, DP081, DP082, DP083, DP085, DP086, DP087 and DP088 are the apheresis group, and DP119 is the peripheral blood group.
Example 3
And (3) virus infection step:
1. collecting T cells after 24-48 hours of activation according to the proportion of 1.5 multiplied by 106cells/mL were plated into culture flasks.
2. The amount of virus required was calculated as multiplicity of infection (MOI).
3. The desired virus volume was aspirated, added to the cell suspension, gently mixed, placed in a carbon dioxide incubator (37 ℃, 5% CO)2) And (5) culturing.
In this protocol, the inventors found that the use of viral infection co-factors did not significantly contribute to CAR expression rates (table 2 and figure 4). Therefore, the invention does not add virus infection accessory factors.
Different infection times have more significant effects on CAR expression rate (Table 3), wherein the virus infection is better than 24 hours after activation, and the virus infection is carried out 24-48 hours after T activation, so as to meet the requirements of clinical production.
As can be seen from the effect of the number of infections on the CAR expression rate, the use of one infection was superior to the second (Table 4), and thus the present invention used a one infection protocol.
Through the adjustment, the operation process of virus infection is simplified, and the risk is reduced.
TABLE 2
Figure BDA0002051682030000122
Figure BDA0002051682030000131
TABLE 3
Figure BDA0002051682030000132
TABLE 4
Figure BDA0002051682030000133
Example 4 cell proliferation
In the cell proliferation process, the cell is cultured by adopting a serum-free and antibiotic-free X-VIVO15 culture medium, so that the dependence of the cell on serum factors and the antibiotic effect in the culture process are reduced, the finished product preparation is free of serum and antibiotic residues, and the safety is ensured. The culture time is shortened to 7-8 days, the clinical controllability is improved, the cells are in a high-speed proliferation stage during the preparation of finished products, and the activity of the cells is ensured. IL-2 (concentration 50IU/mL, 200IU/mL, 500IU/mL) is added in the cell proliferation process.
Cell expansion experiment procedure:
1. 24 hours after infection with virus, cell suspensions were collected and centrifuged (400g/5 min/L8 down 8).
2. The supernatant was discarded, and the medium was adjusted to 0.5X 10 using X-VIVO156cells/mL,0.6×106cells/mL,0.7×106cells/mL,0.8×106cells/mL weightAnd (5) suspension culture.
3. And (4) changing the culture medium every other day, wherein the ratio of the new culture medium to the old culture medium is 5:1, 4:1, 3:1, 2:1 or 1: 1.
4. And collecting cells when the cells are cultured to 7 days or 8 days for finished product preparation.
The effect of different concentrations of IL-2 on cell proliferation is shown in FIG. 5.
The number of viable cells of three patients cultured with CAR-T cells for 7-8 days is shown in FIG. 6.
Finally, it should be noted that: the above embodiments are only used to illustrate the technical solution of the present invention, and not to limit the same; while the invention has been described in detail and with reference to the foregoing embodiments, it will be understood by those skilled in the art that: the technical solutions described in the foregoing embodiments may still be modified, or some or all of the technical features may be equivalently replaced; and the modifications or the substitutions do not make the essence of the corresponding technical solutions depart from the scope of the technical solutions of the embodiments of the present invention.

Claims (10)

1. The method for enriching the T cells is characterized by comprising the steps of culturing Peripheral Blood Mononuclear Cells (PBMCs) for 1-4 hours, removing adherent cells, and separating the T cells from the rest cells by using magnetic beads.
2. The method of claim 1, wherein the T cells selected are CD3+CD28+A T cell;
preferably, the magnetic beads are CD3/CD28 antibody conjugated magnetic beads which are bound to the remaining cells in a ratio of magnetic beads: incubating the T cells according to the proportion of (1-3) to 1, and separating the T cells by using the magnetic beads;
preferably, the seeding density of the PBMCs during culture is (0.5-1.5) × 108cells/30mL;
Preferably, the sorting is performed at a cell density of (0.15 to 0.6). times.106cells/mL;
Preferably, the PBMCs are from a subject in need of adoptive T cell therapy;
preferably, the PBMCs are from a single blood sample, peripheral blood whole blood or peripheral blood stem cells;
preferably, the PBMCs are freshly isolated cells or cells that are thawed after cryopreservation.
3. Use of the method of claim 1 or 2 for the manufacture of a medicament for T cell immunotherapy;
the T cell immunotherapy drug comprises one or more of the following cells: tumor infiltrating lymphocytes, cytotoxic T cells, chimeric antigen receptor T cells (CAR-T), T cell receptor chimeric T cells, natural killer T cells, and peripheral blood lymphocytes.
4. A method of making a CAR-T cell, comprising the steps of:
i) enriching T cells using the method of claim 1 or 2;
ii) inoculating the virus carrying the CAR expression fragment 24-48 hours after T cell activation; and
iii) culturing the T cells.
5. The method of claim 4, wherein the virus carrying the CAR-expressing fragment is a retrovirus;
preferably, the virus carrying the CAR expression fragment is a lentivirus;
preferably, the virus carrying the CAR expression fragment is inoculated only once;
preferably, no infection co-factors are added during the process of making the CAR-T cells.
6. The method according to claim 5, wherein in step iii) the cells are cultured using serum-free antibiotic-free medium.
7. The method according to claim 6, wherein the culture medium contains 100-600 IU/mL of interleukin-2.
8. The method according to claim 6 or 7, wherein in step iii) the seeding density of T-cells is 0.5-0.8 x 106cells/mL。
9. The method as claimed in claim 8, wherein in step iii), the medium is changed every 12-36 hours, and the ratio of new medium to old medium is (5-1): 1.
10. The method according to claim 9, wherein in step iii), the cultivation time is 7 to 8 days in total.
CN201910375997.1A 2019-05-07 2019-05-07 Method for enriching T cells and application of method in adoptive T cell therapy Pending CN111909899A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910375997.1A CN111909899A (en) 2019-05-07 2019-05-07 Method for enriching T cells and application of method in adoptive T cell therapy

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910375997.1A CN111909899A (en) 2019-05-07 2019-05-07 Method for enriching T cells and application of method in adoptive T cell therapy

Publications (1)

Publication Number Publication Date
CN111909899A true CN111909899A (en) 2020-11-10

Family

ID=73242468

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910375997.1A Pending CN111909899A (en) 2019-05-07 2019-05-07 Method for enriching T cells and application of method in adoptive T cell therapy

Country Status (1)

Country Link
CN (1) CN111909899A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023044488A1 (en) * 2021-09-20 2023-03-23 Immatics US, Inc. Monocyte depletion of t cells populations for t-cell therapy

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108660113A (en) * 2018-05-21 2018-10-16 杭州启澜生物医学技术有限公司 A method of preparing modified form CAR-T cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108660113A (en) * 2018-05-21 2018-10-16 杭州启澜生物医学技术有限公司 A method of preparing modified form CAR-T cells

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023044488A1 (en) * 2021-09-20 2023-03-23 Immatics US, Inc. Monocyte depletion of t cells populations for t-cell therapy

Similar Documents

Publication Publication Date Title
CN112626018A (en) High-purity allogeneic NK cell culture medium and in-vitro amplification method
CN113151168B (en) Human NK cell culture system and preparation method thereof
CN104395461B (en) Method for obtaining monocytes or NK cells
WO2006012359A2 (en) Methods for inducing the differentiation of blood monocytes into functional dendritic cells
CN108251369B (en) Immune cell culture medium, culture method and application
CN111117961B (en) Tumor antigen loaded DC-CIK cell culture method
CN110607276A (en) Serum-free culture method for efficiently amplifying cord blood NK cells
CN112852728B (en) LCL-NK cell combined culture method based on peripheral blood, cell and product
CN111909899A (en) Method for enriching T cells and application of method in adoptive T cell therapy
CN114402065A (en) Low density cell culture
CN109535241B (en) DC-CIK (dendritic cell-cytokine induced killer) co-culture cell, preparation method thereof, sensitizing antigen and application
CN112300988A (en) T cell sorting preparation technology based on tumor neoantigen and application thereof
CN110862962A (en) Method for culturing and amplifying NK cells in vitro by using gallic acid
CN110713979B (en) Culture method of CD34+ hematopoietic stem cells
JP2002171966A (en) Activated lymphocyte originating from cord blood, medical preparation containing the same as a main component, method of preparing the same and a kit for preparing the preparation
CN110747167B (en) Preparation method and application of hemizygous BAK cell
CN108441473B (en) Method for enriching CD8+ T cells in vitro
US8313945B2 (en) Methods for inducing the differentiation of blood monocytes into functional dendritic cells
CN113980901B (en) Method for preparing high-purity mature human dendritic cells and application
CN114958740B (en) Method for in vitro culture and enrichment of human NK cells
CN115232789B (en) Method for preparing cytokine-induced killer cells by using Daudi cell membrane capsules
Van Riet et al. Persistence of residual tumour cells after cytokine‐mediated ex vivo expansion of mobilized CD34+ blood cells in multiple myeloma
CN109679908B (en) Method for preparing high-purity human dendritic cells and application thereof
CN117210404A (en) High-purity peripheral blood autologous NK cell culture medium and in-vitro culture method
CN114657124A (en) Preparation method of compound immune cells with high killing capacity on tumor cells

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
RJ01 Rejection of invention patent application after publication

Application publication date: 20201110

RJ01 Rejection of invention patent application after publication