WO2023096352A1 - Lignée de cellules nourricières génétiquement modifiées pour exprimer hla-e et son utilisation - Google Patents

Lignée de cellules nourricières génétiquement modifiées pour exprimer hla-e et son utilisation Download PDF

Info

Publication number
WO2023096352A1
WO2023096352A1 PCT/KR2022/018628 KR2022018628W WO2023096352A1 WO 2023096352 A1 WO2023096352 A1 WO 2023096352A1 KR 2022018628 W KR2022018628 W KR 2022018628W WO 2023096352 A1 WO2023096352 A1 WO 2023096352A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
hla
feeder
cell
genetically engineered
Prior art date
Application number
PCT/KR2022/018628
Other languages
English (en)
Korean (ko)
Inventor
조덕
판티민창
고승권
김현영
Original Assignee
사회복지법인 삼성생명공익재단
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
Priority claimed from KR1020220118160A external-priority patent/KR20230078501A/ko
Application filed by 사회복지법인 삼성생명공익재단 filed Critical 사회복지법인 삼성생명공익재단
Publication of WO2023096352A1 publication Critical patent/WO2023096352A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • 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

Definitions

  • It relates to a feeder cell line genetically engineered to express HLA-E, and its use, specifically, a method for proliferating NK cells using the same, or a method for measuring NK cell activity using the same.
  • NK cells are a type of cytotoxic lymphocytes that are important for innate immunity. NK cells respond to virus-infected cells or cancer cells, and these responses are signals generated by various activating receptors and inhibitory receptors of NK cells reacting with each ligand of target cells. ) depends on Normally, NK cells can attack target cells if the activation signal is stronger than the inhibitory signal, but do not attack if the inhibitory signal is stronger. Therefore, normal cells are not attacked by NK cells because they generate inhibitory signals due to the presence of NK cell inhibitory receptor ligands (MHC, major histocompatibility complex).
  • MHC NK cell inhibitory receptor ligands
  • NK cells may decrease due to the occurrence of MHC abnormalities on the cell surface.
  • NK cells attack target cells. It has cytotoxicity by secreting perforin to pierce the cell membrane of infected cells or cancer cells and releasing granzyme to kill these cells.
  • B-cell lymphoma and many cancer patients defects in the number or anticancer activity of NK cells have been found, and it is known that NK cell dysfunction is closely related to the occurrence of these cancers. Therefore, adoptive cell therapy (ACT), in which a large amount of high-performance NK cells derived from the peripheral blood of healthy people are cultured in vitro and then administered to cancer patients by maximizing anticancer activity, is emerging.
  • ACT adoptive cell therapy
  • the treatment is Development of an amplification method is essential to secure a large amount of high-performance NK cells.
  • NK cell proliferation methods were methods for NK cell (conventional NK cell) amplification.
  • NK cell conventional NK cell
  • adaptive NK cells which are one of the subtypes of NK cells involved in the adaptive immune system, has recently been emphasized as specialized natural killer cells capable of forming immunological memory. Except for one study that showed amplification (Cancer Immunol. Res. 2017, 5, 654-665.), there is no condition.
  • One aspect is to provide feeder cells for culturing NK cells genetically engineered to express human leukocyte antigen (HLA).
  • HLA human leukocyte antigen
  • Another aspect is to provide a composition for NK cell culture comprising the culture feeder cells.
  • Another aspect includes obtaining a blood sample containing a population of NK cells; and contacting at least a portion of the mixed population of NK cells with cells genetically engineered to activate NK cells, wherein the genetically engineered cells are strained to express Human leukocyte antigen (HLA). It is to provide a method of proliferating NK cells, comprising steps that are wholly engineered.
  • HLA Human leukocyte antigen
  • One aspect provides a cell line genetically engineered to express human leukocyte antigen (HLA).
  • HLA human leukocyte antigen
  • the cell line may be a culture feeder cell that selectively amplifies only NK cells.
  • the term "feeder cells (feeder cells)” does not have the ability to divide and proliferate by irradiation, but because it has metabolic activity, it produces various metabolites to help the proliferation of target NK cells. can do.
  • the feeder cell that can be used in the present specification is an animal cell line into which a gene has been introduced, and may be a human chronic myelogenous leukemia cell line (eg, K562 cell), RPMI8866, EBV_LCL, HFWT, and the like.
  • K562 cell line which is a representative nutritional helper cell used for NK cell proliferation, is only used for NK cell proliferation, the K562 cell line itself should not proliferate.
  • the K562 cell line is pretreated by irradiation with strong radiation (eg, 50 to 100 Gy), so that the K562 cell line itself does not proliferate at all, but to help only the proliferation of NK cells do.
  • strong radiation eg, 50 to 100 Gy
  • most cytokines such as IL-2 and IL-15 must be continuously exposed to NK cells. Therefore, when cancer cell line-based feeder cells are genetically engineered to express cytokines such as IL-2 and/or IL-15, the cancer cell line pretreated with radiation or the like cannot survive for a long time in the culture process. There is a problem that the selective proliferation efficiency of NK cells is low.
  • NK cells can be obtained selectively and more efficiently by genetically engineering the cell line according to one aspect to express human leukocyte antigen.
  • the NK cells may be adaptive NK cells.
  • adaptive NK cells are specialized natural killer cells capable of forming an immunological memory and are involved in the adaptive immune system.
  • the adaptive NK cells do not have antigen specificity, survive longer in vivo among NK cells, and have a stronger cell subtype (Ab-dependent cell-mediated cytotoxicity, ADCC), etc. subgroup).
  • the NK cells may be NKG2C+NK cells.
  • the term "Human leukocyte antigen (HLA)” is a cell membrane glycoprotein molecule expressed on the surface of human nucleated cells, which presents an antigen to T lymphocytes to induce an adaptive immune response against the invading antigen. and protects normal cells from apoptosis by NK cells.
  • the HLA may be, for example, HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, HLA-G, and the like.
  • the HLA-E is about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 92% or more of the nucleic acid sequence of SEQ ID NO: 1 or its amino acid sequence. or more, about 95% or more, about 97% or more, about 98% or more, or about 99% or more sequence homology.
  • genetic engineering or “genetically engineered” refers to the act of introducing one or more genetic modifications to a cell or a cell produced thereby. .
  • the genetically engineered cell may contain an exogenous gene encoding the aforementioned gene.
  • exogenous means that the referenced molecule or referenced activity has been introduced into a host cell.
  • a molecule may be introduced as non-chromosomal genetic material such as a plasmid or introduction of an encoding nucleic acid into host genetic material, such as, for example, by insertion into a host chromosome.
  • exogenous refers to introduction of the coding nucleic acid into a subject in a form capable of being expressed.
  • biosynthetic activity the term “exogenous” refers to an activity introduced into the host parent cell.
  • the source may be, for example, a homologous or heterologous encoding nucleic acid that expresses the activity mentioned after being introduced into the parental cell of the host. Therefore, the term “endogenous” refers to the mentioned molecule or activity present in the host cell. Similarly, with respect to expression of an encoding nucleic acid, the term “endogenous” refers to the expression of an encoding nucleic acid contained within a subject.
  • heterologous refers to a molecule or activity from a source other than the species mentioned and the term “homologous” refers to a molecule or activity from the host parental cell.
  • exogenous expression of an encoding nucleic acid may utilize either or both heterologous or homologous encoding nucleic acids.
  • the cell may contain a nucleic acid encoding HLA. More specifically, the cells may be transformed with a vector containing a nucleic acid encoding HLA.
  • the term "vector” refers to a vector capable of expressing a protein of interest in a suitable host cell, and refers to a genetic construct comprising regulatory elements operably linked to express a gene insert.
  • a vector may include expression control elements such as a promoter, an operator, an initiation codon, a stop codon, a polyadenylation signal, and/or an enhancer, and the promoter of the vector may be constitutive or inducible.
  • the vector may be an expression vector capable of stably expressing the fusion protein in a host cell.
  • the expression vector conventional ones used in the art to express foreign proteins in plants, animals, or microorganisms may be used.
  • the recombinant vector may be constructed through various methods known in the art.
  • the vector may include a selectable marker for selecting a host cell containing the vector and, in the case of a replicable vector, an origin of replication.
  • the vector can replicate autonomously or be introduced into host DNA, wherein the vector is selected from the group consisting of plasmids, lentiviruses, adenoviruses, adeno-associated viruses, retroviruses, herpes simplex viruses, and vaccinia viruses. it could be
  • the polynucleotide sequence encoding the aforementioned fusion protein may be operably linked to a promoter.
  • operably linked refers to a functional linkage between a nucleic acid expression control sequence (eg, a promoter, signal sequence, or array of transcriptional regulator binding sites) and another nucleic acid sequence, whereby The regulatory sequence will control the transcription and/or translation of the other nucleic acid sequence.
  • compositions for culturing NK cells comprising cells genetically engineered to express human leukocyte antigen (HLA).
  • HLA human leukocyte antigen
  • composition for NK cell culture comprising cells genetically engineered to express HLA according to one embodiment can induce the amplification and/or activation of NK cells (eg, natural killer cells), thereby culturing NK cells, It can be usefully used for isolation or proliferation.
  • NK cells eg, natural killer cells
  • Another aspect includes obtaining a blood sample containing a population of mixed immune cells; and contacting at least a portion of the mixed population of NK cells with cells genetically engineered to activate NK cells, wherein the genetically engineered cells are strained to express Human leukocyte antigen (HLA).
  • HLA Human leukocyte antigen
  • the contacting step is co-cultivating the population of NK cells mixed with the genetically engineered cells to stimulate, activate, or expand the subpopulation of the NK cells. It may contain.
  • stimulation of NK cells refers to increasing the activity of natural killer cells, for example, cytotoxic activity, in vitro or in vivo, or generating, increasing, amplifying, or proliferating activated natural killer cells. that can mean
  • non-limiting examples of the NK cells include macrophages, B lymphocytes, T lymphocytes, mast cells, monocytes, dendritic cells, eosinophils, natural killer cells, basophils, and neutrophils.
  • the NK cells are selected from the group consisting of macrophages, B lymphocytes, T lymphocytes (CD8+ CTL), mast cells, monocytes, dendritic cells, eosinophils, natural killer cells, basophils, or neutrophils. It could be any one.
  • NK cells may be natural killer cells or T lymphocytes.
  • the mixed NK cells may include one or more selected from the group consisting of macrophages, B lymphocytes, T lymphocytes, mast cells, monocytes, dendritic cells, eosinophils, natural killer cells, basophils, and neutrophils.
  • NK cells are cytotoxic lymphocytes constituting a major component of the innate immune system, and are defined as large granular lymphocytes (LGL) and lymphoid progenitor cells ( Common lymphoid progenitor (CLP) constitutes a third cell differentiated from B and T lymphocytes.
  • LGL large granular lymphocytes
  • CLP Common lymphoid progenitor
  • the "natural killer cells” or “NK cells” include natural killer cells without additional modification derived from any tissue source, and may include mature natural killer cells as well as natural killer progenitor cells.
  • Natural killer cells are activated in response to interferon or macrophage-derived cytokines, and natural killer cells are labeled with "activating receptors" and "inhibitory receptors", which control the cytotoxic activity of cells. Including surface receptors.
  • Natural killer cells can be generated from hematopoietic cells, eg, hematopoietic stems or precursors, from any source, eg, placental tissue, placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver, and the like.
  • the natural killer cells may be activated natural killer cells.
  • the activated natural killer cells may refer to cells in which cytotoxicity or natural killer cells' innate immunomodulatory ability is activated compared to parental cells, eg, hematopoietic cells or natural killer progenitor cells.
  • activated natural killer cells or populations enriched in activated natural killer cells are characterized by one or more functionally relevant markers, such as CD16, CD57, CD69, CD94, CD161, CD158a, CD158b, NKp30. , NKp44, NKp46, DNAM-1, 2B4, NKp46, CD94, KIR (eg KIR2DL1, KIR2DL2/3, KIR3DL1), and the NKG2 family of activating receptors (eg NKG2A, NKG2C, NKG2D) can be evaluated.
  • functionally relevant markers such as CD16, CD57, CD69, CD94, CD161, CD158a, CD158b, NKp30.
  • the co-culture may be performed in the presence of cytokines.
  • cytokine may refer to a protein ( ⁇ 5-20 kDa) that plays a role in cell signaling. Cytokines are released by cells and affect the behavior of cells that release cytokines and/or other cells. Non-limiting examples of cytokines include chemokines, interferons, interleukins, lymphokines, tumor necrosis factors, monokines, and colony stimulating factors. Cytokines can be produced by a wide variety of cells including, but not limited to, immune cells such as macrophages, B lymphocytes, T lymphocytes, mast cells and monocytes, endothelial cells, fibroblasts and stromal cells. can Cytokines can be produced by more than one type of cell.
  • Cytokines act through receptors and are of particular importance in the immune system, regulating the balance between humoral and cellular immune responses, and regulating the maturation, growth and responsiveness of cell populations.
  • a cytokine herein may be a naturally occurring cytokine or may be a mutant version of a naturally occurring cytokine.
  • naturally occurring may also be referred to as wild type and includes allelic variants.
  • a mutated version or "mutation" of a naturally occurring cytokine refers to certain mutations made to a naturally occurring sequence to alter the function, activity and/or specificity of a cytokine.
  • mutations can enhance the function, activity and/or specificity of a cytokine.
  • the mutations can reduce the function, activity and/or specificity of the cytokine.
  • a mutation may include deletion or addition of one or more amino acid residues of a cytokine.
  • the cytokines include BMP (Bone morphogenetic protein) family, CCL (Cheomkine ligands) family, CMTM (CKLF-like MARVEL transmembrane domain containing member) family, CXCL (CXC motif ligand ligand) family, GDF (Growth/differentiation factor) family, Growth hormone, IFN (Interferon) family, IL (Interleukin) family, TNF (Tumor necrosis factors) family, GPI (glycophosphatidylinositol), SLUPR-1 (Secreted Ly-6/uPAR -Related Protein 1), SLUPR-2 (Secreted Ly -6/uPAR -Related Protein 2) and combinations thereof.
  • BMP Bone morphogenetic protein
  • CCL Cheomkine ligands
  • CMTM CKLF-like MARVEL transmembrane domain containing member
  • CXCL CXC motif ligand ligand
  • the cytokine may be an interleukin or a mutant thereof.
  • Many interleukins are synthesized by helper CD4 T lymphocytes, as well as monocytes, macrophages, and endothelial cells. Interleukins can promote the development and differentiation of T and B lymphocytes and hematopoietic cells.
  • Interleukins include, for example, IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8 (CXCL8), IL9, IL10, IL11, IL12, IL13, IL14, IL15, IL16, IL17, IL18, IL19, IL20, IL21 , IL22, IL23, IL24, IL25, IL26, IL27, IL28, IL29, IL30, IL31, IL32, IL33, IL35, or IL36.
  • the cytokine is IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8 (CXCL8), IL9, IL10, IL11, IL12, IL13, IL14, IL15, IL16, IL17, IL18 , including but not limited to wildtype and mutant forms of IL19, IL20, IL21, IL22, IL23, IL24, IL25, IL26, IL27, IL28, IL29, IL30, IL31, IL32, IL33, IL35, or IL36 It may be an interleukin or a mutant thereof.
  • the cytokines added to the co-culture may be IL-2 and IL-15.
  • the IL-2 may be used at a concentration of 1 to 20 ng/ml, 1 to 15 ng/ml, 1 to 10 ng/ml, or 2 to 8 ng/ml.
  • the IL-15 is 10 to 200 ng/mL, 10 to 180 ng/mL, 20 to 160 ng/mL, 20 to 120 ng/mL, 20 to 40 ng/mL, 20 to 80 ng/mL, 40 to 160 ng/ml, 40 to 120 ng/ml, 40 to 80 ng/ml, 80 to 160 ng/ml, 80 to 160 ng/ml, or 80 to 120 ng/ml.
  • the proliferation of NK cells can be further increased synergistically.
  • the co-culture may be performed for 2 to 100 days.
  • the genetically engineered cells may be treated with 50 Gy to 300 Gy radiation.
  • the sample may be a biological sample derived from an individual, for example, a mammal including a human.
  • the biological sample may be isolated from an individual, and may be blood, whole blood, serum, plasma, lymph fluid, urine, feces, tissue, cell, organ, bone marrow, saliva, sputum, cerebrospinal fluid, or a combination thereof.
  • the biological sample may include PBMCs, purified NK cells, or primary resting cells (ie, immediately isolated from blood).
  • Another aspect includes obtaining a blood sample containing a mixed population of NK cells; activating NK cells by contacting at least a portion of the mixed population of NK cells with cells genetically engineered to activate NK cells, wherein the genetically engineered cells have a human leukocyte antigen (HLA) Step that is genetically engineered to express; And it provides a method for examining the activity of NK cells comprising the step of analyzing the degree of activation of the activated NK cells.
  • HLA human leukocyte antigen
  • Another aspect includes obtaining a blood sample containing a mixed population of NK cells; activating NK cells by contacting at least a portion of the mixed population of NK cells with cells genetically engineered to activate NK cells, wherein the genetically engineered cells have a human leukocyte antigen (HLA) Step that is genetically engineered to express; And it provides a method for diagnosing a NK cell-related disease comprising analyzing the degree of activation of the activated NK cells or a method for providing information on the diagnosis.
  • HLA human leukocyte antigen
  • the step of comparing the activation of the NK cells compared to normal NK cells is, specifically, the above-mentioned genetically engineered cells under conditions equivalent to the experimental group using normal NK cells as a control group
  • the activation phenomenon in normal NK cells when stimulated is remarkably high in the experimental group, when it does not appear or when the degree is remarkably low, it is judged as abnormal.
  • pathological signs of diseases related to abnormal NK cells, viral infection, presence of cancer cells, and specific cancers can be determined, and the prognosis of these diseases can be predicted.
  • the "normal NK cell” refers to NK cells possessed by or derived from an individual without the disease, and the individual without the disease is at least a physical, genetic, or exogenous factor known to affect NK cell activity. have no conditions
  • the method may further include isolating required NK cells from the sample.
  • the step of stimulation may be performed in the presence of other blood cells or lymphocytes, and the step of stimulating may be performed after the step is performed to obtain a sample containing only NK cells as lymphocytes.
  • the degree of purification of the isolated NK cells and the composition of the sample may vary to the extent necessary for the experiment. NK cells can be used as they are purified from the sample, if necessary, or can be used after proliferating to secure conditions or cell mass suitable for the experiment.
  • the step of isolating the NK cells may not be essential.
  • measuring the degree of activation may include at least one selected from among degranulation activity, cytotoxicity activity, and measurement of cytokines secreted by NK cell stimulation.
  • the degranulation activity may mean, for example, induction of lysis of target cells by secretion of perforin or granzyme, and this may be analyzed using FACS.
  • FACS Fluorescence Activated Cell Sorting
  • the cytotoxic activity for example, after culturing with a culture containing target cells capable of activating NK cells labeled with europium fluorescent dye, the amount of fluorescent dye released through target cell lysis It can be measured using a microplate reader.
  • the cytokine may be one selected from IFN- ⁇ , TNF- ⁇ , TNF- ⁇ , MIP-1 ⁇ , MIP-1 ⁇ , PANTES, IL-8 and IL-10.
  • Analysis of immunoactivator expression of NK cells as described above may be performed using FACS, intracellular cytokine staining, or ELISA. Specifically, after staining the NK cell surface using a specific fluorochrome-conjugated antibody, the cells are permeabilized (permeabilization), and another specific fluorochrome-conjugated immunoactivator (eg, IFN- ⁇ antibody)
  • a method of measuring the expression of immunoactivating factors in NK cells by staining cytokines and the like can be used.
  • the disease associated with the activity of the NK cell is one that shows abnormal NK cell activity, for example, hyperimmune disease, autoimmune disease, immune rejection, immunodeficiency disease, histiocytosis, cancer, 2 Type 2 diabetes, parasitic infections and viral diseases.
  • the hypersensitive immune disease is at least one selected from asthma and empyema
  • the autoimmune disease is at least one selected from lupus, multiple sclerosis, type 1 diabetes and rheumatoid arthritis
  • the histiocytosis is HLH, XLP1 , and may be any one or more selected from XLP2.
  • Hemophagocytic lymphohistiocytosis is characterized by group 2 Langerhans cell histiocytosis, erythrophagocytic lymphohistiocytosis (familial, sporadic), infection-associated hemophagocytic syndrome, virus-associated hemophagocytic syndrome, and giant lymphadenopathy. Histiocytosis, or reticular histiocytosis.
  • the "cancer” is meant to include tumor, hematological cancer, or solid cancer, and includes those that impair the synergistic activity of NK cells in an individual or do not cause synergistic activity of NK cells as target cells under specific conditions.
  • the cancer is lung cancer, liver cancer, esophageal cancer, stomach cancer, colon cancer, small intestine cancer, pancreatic cancer, melanoma, breast cancer, oral cancer, brain tumor, thyroid cancer, parathyroid cancer, kidney cancer, cervical cancer, sarcoma, prostate cancer, urethra It may be selected from the group consisting of cancer, bladder cancer, testicular cancer, blood cancer, lymphoma, skin cancer, psoriasis, and fibroadenoma.
  • the cancer may be pancreatic cancer or B cell lymphoma.
  • the viral disease may be hepatitis B.
  • the immunodeficiency disease may be DiGeorge syndrome or Chedial-Higashi syndrome.
  • Information on the disease related to NK cell activity can be determined as abnormal NK cells when activation of NK cells in the experimental group is not detected compared to normal NK cells, or NK cells that have lost activity for a specific receptor are abnormal for target cells. If it causes or does not cause activity as a result, it can be determined that the target cell is related to a specific disease.
  • NK cells prepared by the method for propagating NK cells.
  • the NK cells may be genetically engineered to express human leukocyte antigen (HLA).
  • HLA human leukocyte antigen
  • Another aspect provides a cell therapy agent comprising the immune cells or cell population thereof as an active ingredient.
  • Another aspect provides a pharmaceutical composition for preventing or treating cancer or infectious disease using the immune cells or cell population thereof as an active ingredient.
  • Another aspect provides the use of the immune cells or cell population thereof for the manufacture of a medicament.
  • Another aspect provides a method for treating a disease comprising administering the immune cells or cell population thereof to a subject.
  • disease may mean one pathological condition, particularly cancer, infectious disease, inflammatory disease, metabolic disease, autoimmune disorder, degenerative disease, apoptosis-related disease, and graft rejection.
  • treatment refers to, includes, or alleviates, inhibits the progress of, or prevents a disease, disorder or condition, or one or more symptoms thereof, and an "active ingredient” or “pharmaceutically effective amount” refers to a disease, disorder or condition. , or any amount of a composition used in the practice of the invention provided herein sufficient to alleviate, inhibit the progression of, or prevent one or more symptoms thereof.
  • administering As used herein, the terms “administering,” “introducing,” and “implanting” are used interchangeably and according to one embodiment, the administration of a composition into a subject by a method or route that results in at least partial localization to a desired site. It may refer to the arrangement of a composition according to one embodiment. It can be administered by any suitable route that delivers at least a portion of the cells or cellular components of a composition according to one embodiment to a desired location within a living subject.
  • the survival period of the cells after administration to the subject may be as short as several hours, for example, 24 hours to several days, or as long as several years.
  • isolated cell eg, "isolated immune cell” and the like, refers to a cell substantially separated from a tissue of origin, eg, hematopoietic cell.
  • the method of administering the pharmaceutical composition is not particularly limited, but may be administered orally or parenterally, such as intravenous, subcutaneous, intraperitoneal, inhalation or topical application, depending on the desired method.
  • the dosage varies depending on the patient's weight, age, sex, health condition, diet, administration time, administration method, excretion rate, and severity of the disease.
  • a daily dose refers to an amount of a therapeutic substance according to one aspect sufficient to treat a disease state alleviated by administration to a subject in need thereof.
  • An effective amount of a therapeutic agent will depend on the particular compound, the disease state and its severity, and the subject in need of treatment, and can be routinely determined by one skilled in the art.
  • the dosage of the composition according to one aspect to the human body may vary depending on the patient's age, weight, sex, dosage form, state of health, and degree of disease. Based on an adult patient weighing 70 kg, for example, about 1,000 to 10,000 cells/time, 1,000 to 100,000 cells/time, 1,000 to 1000,000 cells/time, 1,000 to 10,000,000, 1,000 to 100,000,000 cells/time, 1,000 to 1,000,000,000 cells/time, 1,000 to 10,000,000,000 cells/circuit, once or several times a day at regular time intervals, divided administration may be administered, or multiple times at regular time intervals.
  • 'Individual' means a subject in need of treatment for a disease, and more specifically, means a mammal such as a human or non-human primate, mouse, rat, dog, cat, horse, and cow. .
  • a pharmaceutical composition according to one embodiment may include a pharmaceutically acceptable carrier and/or additives.
  • a pharmaceutically acceptable carrier and/or additives For example, sterile water, physiological saline, common buffers (phosphoric acid, citric acid, other organic acids, etc.), stabilizers, salts, antioxidants (ascorbic acid, etc.), surfactants, suspending agents, tonicity agents, or preservatives, etc. can do.
  • it may also include combining organic substances such as biopolymers, inorganic substances such as hydroxyapatite, specifically collagen matrices, polylactic acid polymers or copolymers, polyethylene glycol polymers or copolymers, and chemical derivatives thereof.
  • the pharmaceutical composition according to one embodiment is prepared in a formulation suitable for injection, immune cells, immune cells, or substances that increase their activity are dissolved in a pharmaceutically acceptable carrier or in a dissolved solution state. may be frozen.
  • the pharmaceutical composition according to one embodiment if necessary according to the administration method or dosage form, suspending agent, solubilizing agent, stabilizer, isotonic agent, preservative, anti-adsorption agent, surfactant, diluent, excipient, pH adjuster, analgesic agent, Buffers, reducing agents, antioxidants and the like may be appropriately included.
  • Pharmaceutically acceptable carriers and agents suitable for the present invention including those exemplified above, are described in detail in Remington's Pharmaceutical Sciences, 19th ed., 1995.
  • the pharmaceutical composition according to one embodiment is formulated in unit dosage form by using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily performed by those skilled in the art.
  • the dosage form may be in the form of a solution, suspension or emulsion in an oil or aqueous medium, or in the form of a powder, granule, tablet or capsule.
  • the proliferation and activity of adaptive NK cells can be increased by at least two to several times. It can be proliferated and used as a cell therapy.
  • Figure 1a is a result of analyzing the expression of HLA-E in genetically engineered K562 cells using RT-qPCR.
  • Figure 1b is the result of analyzing the expression of HLA-E using flow cytometry (FACS).
  • Figure 2a is a result of measuring the activation of NK cells amplified by feeder cells according to one aspect.
  • Figure 2b is a graph showing Fold expansion of NK cells amplified by feeder cells according to one aspect.
  • Figure 2c is a graph showing the long-term culture effect of the culture helper cells according to one aspect.
  • Figure 3a is a diagram confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect, specifically confirming the levels of CD16 and CD57 (dark black: K562-HLA-E, gray: K562) .
  • Figure 3b is a diagram confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect, specifically confirming the levels of CD69 and NKp30 (dark black: K562-HLA-E, gray: K562) .
  • Figure 3c is a diagram confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect, specifically, confirming the levels of NKp46 and DNAM-1 (dark black: K562-HLA-E, gray: K562).
  • Figure 3d is a diagram confirming the level of NKG2D and NKG2A as a result of confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect (dark black: K562-HLA-E, gray: K562) .
  • Figure 3e is a diagram confirming the level of FcRy as a result of confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect (dark black: K562-HLA-E, gray: K562).
  • Figure 3f is a graph comparing phenotypic expression levels of NK cells amplified by feeder cells and K562 feeder cells according to one aspect (dark black: K562-HLA-E, gray: K562) .
  • Figure 3g is a graph comparing the phenotypic expression levels of NK cells amplified by feeder cells and K562 feeder cells according to one aspect in terms of mean fluorescence intensity (MFI) (dark black : K562-HLA-E, gray: K562).
  • MFI mean fluorescence intensity
  • Figure 4a is a result confirming the Fc ⁇ RI ⁇ -NK cell and NKG2C + NK cell frequency correlation of NK cells amplified by feeder cells and NK cells amplified by K562 cells according to one aspect.
  • Figure 4b is a result of comparing Fc ⁇ RI ⁇ -expression rates of NK cells amplified by feeder cells and NK cells amplified by K562 cells according to one aspect.
  • Figure 4c is a result of confirming Fc ⁇ RI ⁇ cell and NKG2C expression of NK cells amplified by feeder cells and NK cells amplified by K562 cells according to one aspect.
  • Figure 5a is a graph confirming the cytotoxicity (ADCC) of NK cells amplified by feeder cells according to one aspect to K562 cells and Raji cells.
  • Figure 5b is a graph confirming the cytotoxicity according to the HLA-C type of NK cells amplified by feeder cells according to one aspect.
  • Figure 6a is a graph measuring CD107a expression of NK cells amplified by feeder cells according to one aspect.
  • Figure 6b is a graph measuring IFN- ⁇ expression of NK cells amplified by feeder cells according to one aspect.
  • HLA human leukocyte antigen
  • the human-derived HLA-E gene (SEQ ID NO: 1) was cloned into a lentiviral vector, pCDH-CMV-EF1-GFP, to prepare a vector for producing a recombinant lentivirus. Thereafter, the prepared recombinant gene (pCDH-CMV-HLA-E-EF1-GFP) was transfected into 293FT cells together with a packaging vector using lipofectamin3000 (Invitrogen) for virus production. After a period of time, the cells were cultured for 48 hours by replacing the medium with a fresh medium, and then the virus-containing medium was recovered.
  • pCDH-CMV-HLA-E-EF1-GFP lentiviral vector
  • the prepared recombinant gene (pCDH-CMV-HLA-E-EF1-GFP) was transfected into 293FT cells together with a packaging vector using lipofectamin3000 (Invitrogen
  • the recovered medium was centrifuged at 500 ⁇ g for 10 minutes, and only virus-containing pure medium was separated using a 0.45 ⁇ m filter to produce HLA-E-expressing lentivirus. Thereafter, 1 ml of HLA-E expressing lentivirus was dissolved in 9 ml of medium containing K562 cells and added together with polybrene (8 ⁇ g/ml), followed by cell culture for 48 hours. Then, infected cells were selected using RT-qPCR (Quantitative reverse transcription PCR) and flow cytometry (FACS).
  • RT-qPCR Quantitative reverse transcription PCR
  • FACS flow cytometry
  • Figure 1a is a result of analyzing the expression of HLA-E in genetically engineered K562 cells using RT-qPCR.
  • Figure 1b is the result of analyzing the expression of HLA-E using flow cytometry (FACS).
  • the expression level of HLA-E in the genetically engineered K562 cells was significantly higher than that of conventional K562 cells.
  • FIG. 1B it was confirmed that the conventional K562 cells did not express HLA-E, but the genetically engineered K562 cells expressed HLA-E. Therefore, the K562 cell line expressing HLA-E was named "K562-HLA-E".
  • PBMC peripheral blood mononuclear cells
  • the PBMCs were cultured in RPMI 1640 medium containing 10 U/mL recombinant human IL-2 (containing 10% FBS, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin and 4 mmol/L L-glutamine) at 24 - K562 and K562-HLA-E cells (Example 1) irradiated with 100 Gy gamma rays were co-cultured in a well plate, respectively. After 7 days, the IL-2 concentration was increased from 10 U/mL to 100 U/mL, and 5 ng/mL of aqueous IL-5 was added.
  • feeder cells irradiated with gamma rays were added for re-stimulation, and cultured up to 98 days while changing the medium every 2 to 3 days.
  • the expression levels of NKG2C, NKG2A, KIR2DL1, and KIR2DL2/3 were confirmed using flow cytometry (FACS).
  • the expression of NKG2C, KIR2DL1, KIR2DL2/3 and KIR3DL1 was measured according to the number of culture days.
  • the number of NK cells according to the number of culture days was divided by the absolute number of NK cells compared to day 0 and expressed as fold expansion.
  • Figure 2a is a result of measuring the activation of NK cells amplified by feeder cells according to one aspect.
  • Figure 2b is a graph showing Fold expansion of NK cells amplified by feeder cells according to one aspect.
  • Figure 2c is a graph showing the long-term culture effect of the culture helper cells according to one aspect.
  • the feeder cells of Example 1 were significantly higher in NKG2C+ NK cells with self-specific KIR2DL2/3 expression compared to the K562 feeder cells. .
  • NK cells expressing NKG2C, KIR2DL1, KIR2DL2/3 and KIR3DL1 were amplified from day 42 to day 49, but no further amplification was confirmed thereafter. there was.
  • feeder cells of Example 1 it was confirmed that NK cells expressing NKG2C, KIR2DL1, KIR2DL2/3 and KIR3DL1 were amplified even after 90 days.
  • feeder cells not only significantly increase the activity of adaptive NK cells, but also allow long-term culture of NK cells, so that NK cells can be massively proliferated and used as a cell therapeutic agent.
  • the phenotypic characteristics of the NK cells amplified by the feeder cells according to one aspect were confirmed.
  • NK cells amplified in the same manner as in Example 2 with FACS buffer PBS containing 1% FBS
  • FACS buffer PBS containing 1% FBS
  • Cells were then harvested and further stained for 30 minutes with different fluorescently-conjugated anti-human CD16, CD57, CD69, NKp30, NKp46, DNAM-1, NKG2D, NKG2A and FcR ⁇ membrane antibodies, respectively.
  • FACS buffer data were acquired using FACS Verse (BD Biosciences) and analyzed with Kaluza.
  • NK cells amplified by K562 cells were used.
  • Figure 3a is a diagram confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect, specifically confirming the levels of CD16 and CD57 (dark black: K562-HLA-E, gray: K562) .
  • Figure 3b is a diagram confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect, specifically confirming the levels of CD69 and NKp30 (dark black: K562-HLA-E, gray: K562) .
  • Figure 3c is a diagram confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect, specifically, confirming the levels of NKp46 and DNAM-1 (dark black: K562-HLA-E, gray: K562).
  • Figure 3d is a diagram confirming the level of NKG2D and NKG2A as a result of confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect (dark black: K562-HLA-E, gray: K562) .
  • Figure 3e is a diagram confirming the level of FcRy as a result of confirming the phenotypic characteristics of NK cells amplified by feeder cells according to one aspect (dark black: K562-HLA-E, gray: K562).
  • Figure 3f is a graph comparing phenotypic expression levels of NK cells amplified by feeder cells and K562 feeder cells according to one aspect (dark black: K562-HLA-E, gray: K562) .
  • Figure 3g is a graph comparing the phenotypic expression levels of NK cells amplified by feeder cells and K562 feeder cells according to one aspect in terms of mean fluorescence intensity (MFI) (dark black : K562-HLA-E, gray: K562).
  • MFI mean fluorescence intensity
  • the NK cells amplified by the feeder cells of Example 1 had surface markers such as CD16, CD57, CD69, NKp30, NKp46, DNAM-1, NKG2D, NKG2A and FcR ⁇ . expression could be confirmed. Specifically, in the case of NKp30, NKG2A, and NKp46, expression levels were lower than that of NK cells amplified by K562 feeder cells, and CD57, a surface marker of adaptive NK cells, and intracellular receptors In the case of FcR ⁇ , the expression level was higher than that of NK cells amplified by K562 feeder cells.
  • CD16 showed a lower expression level than that of NK cells amplified by K562 feeder cells (see Figs. 3f and 3g). This is thought to be because the adaptive NK cells preferentially produce cytokines including IFN- ⁇ in response to Fc ⁇ RIIIa triggering.
  • NK cells amplified by feeder cells may have complete functional characteristics of adaptive NK cells.
  • Adaptive NK cells have an Fc ⁇ RI ⁇ -deficient phenotype. Therefore, it was confirmed whether the NK cells amplified by the feeder cells according to one aspect also had the same phenotype.
  • NK cells amplified in the same manner as in Example 2 with FACS buffer (PBS containing 1% FBS), APC-Cyanine7-conjugated mouse anti-human CD3 and PE-Cyanine7 conjugated Treatment with anti-human CD56 membrane antibody for 20 minutes. Thereafter, the cells were washed with FACS buffer, permeabilization and fixation were performed to confirm the expression of Fc ⁇ RI ⁇ , an intracellular signal receptor, and then FITC-conjugated mouse anti-human Fc ⁇ RI ⁇ antibody treated for 30 minutes. Thereafter, after washing with FACS buffer, data were acquired using FACS Verse (BD Biosciences) and analyzed with Kaluza.
  • FACS buffer PBS containing 1% FBS
  • Figure 4a is a result confirming the Fc ⁇ RI ⁇ -NK cell and NKG2C + NK cell frequency correlation of NK cells amplified by feeder cells and NK cells amplified by K562 cells according to one aspect.
  • Figure 4b is a result of comparing Fc ⁇ RI ⁇ -expression rates of NK cells amplified by feeder cells and NK cells amplified by K562 cells according to one aspect.
  • Figure 4c is a result of confirming Fc ⁇ RI ⁇ cell and NKG2C expression of NK cells amplified by feeder cells and NK cells amplified by K562 cells according to one aspect.
  • NKG2C positive NK cells on day 0 were confirmed to have a correlation with Fc ⁇ RI ⁇ -, but when the culture was maintained for more than 28 days, it was confirmed that no correlation between NKG2C expression rate and Fc ⁇ RI ⁇ - was observed. there was.
  • cytotoxicity of NK cells on day 14 and day 49 against target cells was measured for 4 hours by CFSE-based assay. Specifically, target cells were stained with 0.5 ⁇ M CFSE for 10 minutes at 37° C. in FACS buffer and washed twice with complete medium.
  • K562 cells stained with CFSE were placed in a 96-well round bottom plate (96-well U-bottom plate) and NK cells amplified by K562-HLA-E and K562 feeder cells
  • the amplified NK cells were mixed at effector-to-target (E:T) ratios of 1:1, 0.5:1, and 0.25:1, respectively, and cultured in a 37°C, 5% CO 2 incubator for 4 hours.
  • E:T effector-to-target
  • CFSE-stained Raji cells were cultured with 1 ⁇ g/ml of rituximab and then cultured in the same manner as above. Thereafter, the mixed cells were transferred to a FACD tube and 1 ⁇ g of 1 mg/ml PI (Invitrogen) was added to each tube, and the cells were acquired in FACS Verse and analyzed using Kaluza software.
  • Figure 5a is a graph confirming direct cytotoxicity of K562 cells on days 14 and 49 of culture of NK cells amplified by feeder cells according to one aspect, and cytotoxicity (ADCC) by addition of Raji cells and Rituximab. .
  • the NK cells amplified by the feeder cells of Example 1 and the NK cells amplified by the K562 feeder cells exhibit similar cytotoxicity to Raji cells bound to K562 and Rituximab. could confirm that
  • NK cells amplified by feeder cells exhibit cytotoxicity and can be used as antibody therapeutics.
  • HLA-C1 and HLA-C2 types were analyzed using the HLA-C SSP PCR kit as a PCR-SSP (PCR amplification with sequence-specific primers) test method.
  • the donor's CMV serology and HLA-C genotype are shown in Table 1 below. Thereafter, cytotoxicity according to the genotype was confirmed.
  • Figure 5b is a graph confirming the cytotoxicity according to the HLA-C type of NK cells amplified by feeder cells according to one aspect.
  • the NK cells amplified by the feeder cells of Example 1 showed improved cytotoxicity against MCF7 cancer cells in C1C1 NK cells compared to NK cells with an HLA-C genotype of C2C2. could confirm that
  • the NK cells amplified by the feeder cells can be usefully used as a therapeutic agent having improved killing efficacy against HLA-C mismatched target cancer cells.
  • the level of CD107a expression on the surface of NK cells proportional to CD107a degranulation of NK cells was measured.
  • NK cells 2 ⁇ 10 5 of NK cells, 2 ⁇ 10 5 of target cells (K562 and MCF7), and 5 ⁇ M of PE-conjugated anti-human CD107a were amplified in the same manner as in Example 2 in a 95-well round-bottom plate. cultured. After 1 hour, Monensin and brefeldin A (BD Biosciences) were added, followed by further incubation for 4 hours. NK cells were then obtained by staining with anti-human CD3 and CD56 antibodies.
  • Figure 6a is a graph measuring CD107a expression of NK cells amplified by feeder cells according to one aspect.
  • NK cells amplified by the feeder cells of Example 1 showed CD107a expression characteristics similar to those of the NK cells amplified by K562 feeder cells or MCF7 target cells.
  • cytotoxicity of NK cells by intracellular IFN- ⁇ production was confirmed.
  • NK cells amplified in the same manner as in Example 2 were cultured in a 96-well round bottom plate at 37° C., 5% CO 2 for 5 hours in the presence of brefeldin A (BD Biosciences) and Monensin (BD Biosciences) did Cells were then harvested and washed by FACS before staining with anti-human CD3 and CD56 membrane antibodies for 20 minutes. After washing, fixation and permeabilization, NK cells were further stained with PE-conjugated anti-human IFN- ⁇ antibody for 30 min on ice. Then, after washing the cells, the cells were obtained in FACS Verse and analyzed using Kaluza software.
  • brefeldin A BD Biosciences
  • Monensin BD Biosciences
  • Figure 6b is a graph measuring IFN- ⁇ expression of NK cells amplified by feeder cells according to one aspect.
  • the NK cells amplified by the feeder cells of Example 1 showed a similar level of IFN- ⁇ expression to that of the NK cells amplified by the K562 feeder cells. That is, it was confirmed that NK cells with increased activity were successfully generated from peripheral blood mononuclear cells of a normal donor.
  • the NK cells amplified by the feeder cells are effective in producing NK cells having high immunoregulatory activity by cytokines.

Landscapes

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

Abstract

La présente invention concerne des cellules génétiquement modifiées pour l'activation des cellules NK. Les cellules génétiquement modifiées pour l'activation des cellules NK selon un aspect peuvent induire de manière synergique la prolifération et l'activation des cellules NK à partir d'un échantillon, et ainsi un procédé de prolifération des cellules NK ou les cellules NK proliférées par le procédé peuvent être utilement employées pour les thérapies par anticorps et autres.
PCT/KR2022/018628 2021-11-25 2022-11-23 Lignée de cellules nourricières génétiquement modifiées pour exprimer hla-e et son utilisation WO2023096352A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2021-0164859 2021-11-25
KR20210164859 2021-11-25
KR10-2022-0118160 2022-09-19
KR1020220118160A KR20230078501A (ko) 2021-11-25 2022-09-19 Hla-e를 발현하도록 유전적으로 조작된 배양보조세포주, 및 그의 용도

Publications (1)

Publication Number Publication Date
WO2023096352A1 true WO2023096352A1 (fr) 2023-06-01

Family

ID=86540045

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/018628 WO2023096352A1 (fr) 2021-11-25 2022-11-23 Lignée de cellules nourricières génétiquement modifiées pour exprimer hla-e et son utilisation

Country Status (1)

Country Link
WO (1) WO2023096352A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180057641A (ko) * 2015-09-04 2018-05-30 밀테니 비오텍 게앰베하 자연살해 세포의 증식방법
US20210355446A1 (en) * 2018-11-23 2021-11-18 Technische Universitat Dresden Artificial hla-positive feeder cell lines for nk cells and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180057641A (ko) * 2015-09-04 2018-05-30 밀테니 비오텍 게앰베하 자연살해 세포의 증식방법
US20210355446A1 (en) * 2018-11-23 2021-11-18 Technische Universitat Dresden Artificial hla-positive feeder cell lines for nk cells and uses thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BJÖRKSTRÖM NIKLAS K., LINDGREN THERESE, STOLTZ MALIN, FAURIAT CYRIL, BRAUN MONIKA, EVANDER MAGNUS, MICHAËLSSON JAKOB, MALMBERG KAR: "Rapid expansion and long-term persistence of elevated NK cell numbers in humans infected with hantavirus", JOURNAL OF EXPERIMENTAL MEDICINE, ROCKEFELLER UNIVERSITY PRESS, US, vol. 208, no. 1, 17 January 2011 (2011-01-17), US , pages 13 - 21, XP093069239, ISSN: 0022-1007, DOI: 10.1084/jem.20100762 *
DUCK CHO, SHIN SHI-WON, PARK JUNG-SUN, KANG HYUN-KYU, KIM SANG-KI, NGUYEN PHAM THAN-NHAN, ZHU XIAO-WEI, SHIN MYUNG-GEUN, SUH SOON-: "Selective Expansion of Natural Killer Cells from Peripheral Blood Mononuclear Cells by K562 Cell Line and IL-2", KOREAN J. HEMATOL., vol. 41, no. 1, pages 8 - 15, XP093069242, DOI: 10.5045/kjh.2006.41.1.8 *
M. SENSI, PIETRA G., MOLLA A., NICOLINI G., VEGETTI C., BERSANI I., MILLO E., WEISS E., MORETTA L., MINGARI M. C., ANICHINI A.: "Peptides with dual binding specificity for HLA-A2 and HLA-E are encoded by alternatively spliced isoforms of the antioxidant enzyme peroxiredoxin 5", INTERNATIONAL IMMUNOLOGY, OXFORD UNIVERSITY PRESS, GB, vol. 21, no. 3, 30 January 2009 (2009-01-30), GB , pages 257 - 268, XP055407526, ISSN: 0953-8178, DOI: 10.1093/intimm/dxn141 *
PHAN MINH-TRANG THI, KIM JINHO, KOH SEUNG KWON, LIM YUREE, YU HONGBI, LEE MIJEONG, LEE JONG-MIN, KANG EUN-SUK, KIM HYUN-YOUNG, KIM: "Selective Expansion of NKG2C+ Adaptive NK Cells Using K562 Cells Expressing HLA-E", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 23, no. 16, pages 9426, XP093069244, DOI: 10.3390/ijms23169426 *

Similar Documents

Publication Publication Date Title
KR101643165B1 (ko) 말초혈액단핵구 유래 자연 살해세포의 유도 및 증식 방법
WO2016085248A1 (fr) Procédé de culture de cellules tueuses naturelles à l'aide de lymphocytes t
WO2017142187A1 (fr) Kit d'addition de milieu pour la culture de cellules nk et procédé de culture de cellules nk (cellules tueuses naturelles) utilisant le kit
US20140348809A1 (en) Compositions and methods for generating interleukin-35-induced regulatory t cells
Abo et al. Extrathymic pathways of T-cell differentiation in the liver and other organs
KR102236011B1 (ko) Nk 세포의 대량증식 배양방법
KR20180054663A (ko) CD8+CD45RClow TREGS의 신규한 아집단 및 이들의 용도
WO2019117633A1 (fr) Composition cosmétique et composition pharmaceutique pour atténuer la dermatite atopique, la chute des cheveux et les plaies ou réduire les rides de la peau
WO2021251707A1 (fr) Lignée cellulaire génétiquement modifiée pour l'activation et l'amplification de cellules nk et utilisation associée
EP1233058B1 (fr) Methode de proliferation de cellules tueuses naturelles
KR101867942B1 (ko) 바이러스 항원 특이적인 t 세포의 유도 및 증식 방법
WO2021251708A1 (fr) Souche cellulaire génétiquement modifiée pour activer et amplifier les cellules nk et son utilisation
WO2017003153A1 (fr) Méthode de production de cellules tueuses naturelles à partir de monocytes de sang de cordon ombilical, ou de cellules dérivées de ces derniers
WO2019231243A1 (fr) Cellule nourricière exprimant ox40l et procédé de culture de cellules tueuses naturelles l'utilisant
WO2023096352A1 (fr) Lignée de cellules nourricières génétiquement modifiées pour exprimer hla-e et son utilisation
KR102596913B1 (ko) Nk 세포의 활성화 및 증폭을 위해 유전적으로 조작된 세포주, 및 그의 용도
US20230302130A1 (en) Poly-donor cd4+ t cells expressing il-10 and uses thereof
KR20230078501A (ko) Hla-e를 발현하도록 유전적으로 조작된 배양보조세포주, 및 그의 용도
WO2023106894A1 (fr) Lymphocytes t de panck restreints par mr1 et leur procédé de fabrication
CN115803436A (zh) 免疫原性降低的新型移植细胞
Nitta et al. Specific CTL activity of CD8+ TCR Vβ14+ T cell in mouse 2, 4, 6-trinitrobenzene sulfonic acid-induced colitis
Nakajima et al. Protective Roles of B and T Lymphocyte
강병현 PLZF+ innate T cells support TGF-b-dependent generation of activated/memory-like regulatory T cells
Baine Helios regulates interleukin-2 expression and suppressive function in natural regulatory T cells
AU2007202851A1 (en) Compositions and methods of monoclonal and polyclonal antibodies specific for T cell subpopulations

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: 22899034

Country of ref document: EP

Kind code of ref document: A1