WO2025022372A1 - Cell population production method - Google Patents
Cell population production method Download PDFInfo
- Publication number
- WO2025022372A1 WO2025022372A1 PCT/IB2024/059123 IB2024059123W WO2025022372A1 WO 2025022372 A1 WO2025022372 A1 WO 2025022372A1 IB 2024059123 W IB2024059123 W IB 2024059123W WO 2025022372 A1 WO2025022372 A1 WO 2025022372A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- medium
- cell
- day
- cell population
- Prior art date
Links
- 238000004519 manufacturing process Methods 0.000 title claims abstract description 39
- 210000004027 cell Anatomy 0.000 claims abstract description 532
- 108010074108 interleukin-21 Proteins 0.000 claims abstract description 134
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 claims abstract description 54
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 claims abstract description 54
- 238000000034 method Methods 0.000 claims abstract description 52
- 239000000556 agonist Substances 0.000 claims abstract description 50
- 108010002350 Interleukin-2 Proteins 0.000 claims abstract description 44
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 claims abstract description 39
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 claims abstract description 39
- 101000581981 Homo sapiens Neural cell adhesion molecule 1 Proteins 0.000 claims abstract description 29
- 102100027347 Neural cell adhesion molecule 1 Human genes 0.000 claims abstract description 29
- 238000012258 culturing Methods 0.000 claims abstract description 18
- 210000005087 mononuclear cell Anatomy 0.000 claims abstract description 15
- 108010082808 4-1BB Ligand Proteins 0.000 claims description 38
- 102100032101 Tumor necrosis factor ligand superfamily member 9 Human genes 0.000 claims description 38
- 239000006166 lysate Substances 0.000 claims description 3
- 239000007788 liquid Substances 0.000 abstract description 5
- 238000012239 gene modification Methods 0.000 abstract 1
- 230000005017 genetic modification Effects 0.000 abstract 1
- 235000013617 genetically modified food Nutrition 0.000 abstract 1
- 239000002609 medium Substances 0.000 description 321
- 239000006285 cell suspension Substances 0.000 description 129
- 102100030704 Interleukin-21 Human genes 0.000 description 128
- 229950005972 urelumab Drugs 0.000 description 100
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 43
- 102000000588 Interleukin-2 Human genes 0.000 description 40
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 39
- 230000001472 cytotoxic effect Effects 0.000 description 29
- QJJXYPPXXYFBGM-LFZNUXCKSA-N Tacrolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1\C=C(/C)[C@@H]1[C@H](C)[C@@H](O)CC(=O)[C@H](CC=C)/C=C(C)/C[C@H](C)C[C@H](OC)[C@H]([C@H](C[C@H]2C)OC)O[C@@]2(O)C(=O)C(=O)N2CCCC[C@H]2C(=O)O1 QJJXYPPXXYFBGM-LFZNUXCKSA-N 0.000 description 28
- 210000000822 natural killer cell Anatomy 0.000 description 28
- 229920000669 heparin Polymers 0.000 description 23
- 229940115586 simulect Drugs 0.000 description 22
- ZFGMDIBRIDKWMY-PASTXAENSA-N heparin Chemical compound CC(O)=N[C@@H]1[C@@H](O)[C@H](O)[C@@H](COS(O)(=O)=O)O[C@@H]1O[C@@H]1[C@@H](C(O)=O)O[C@@H](O[C@H]2[C@@H]([C@@H](OS(O)(=O)=O)[C@@H](O[C@@H]3[C@@H](OC(O)[C@H](OS(O)(=O)=O)[C@H]3O)C(O)=O)O[C@@H]2O)CS(O)(=O)=O)[C@H](O)[C@H]1O ZFGMDIBRIDKWMY-PASTXAENSA-N 0.000 description 19
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 17
- 206010028980 Neoplasm Diseases 0.000 description 17
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 17
- 239000012091 fetal bovine serum Substances 0.000 description 17
- 239000002953 phosphate buffered saline Substances 0.000 description 17
- 229940072288 prograf Drugs 0.000 description 17
- 239000008194 pharmaceutical composition Substances 0.000 description 16
- 239000012980 RPMI-1640 medium Substances 0.000 description 14
- 239000002458 cell surface marker Substances 0.000 description 14
- 108090000623 proteins and genes Proteins 0.000 description 14
- 229920001184 polypeptide Polymers 0.000 description 13
- 102000004196 processed proteins & peptides Human genes 0.000 description 13
- 108090000765 processed proteins & peptides Proteins 0.000 description 13
- 238000012360 testing method Methods 0.000 description 13
- 239000012979 RPMI medium Substances 0.000 description 12
- 230000035755 proliferation Effects 0.000 description 12
- 210000004881 tumor cell Anatomy 0.000 description 12
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 11
- 239000000654 additive Substances 0.000 description 11
- 230000030833 cell death Effects 0.000 description 11
- 238000000684 flow cytometry Methods 0.000 description 11
- 239000000203 mixture Substances 0.000 description 11
- 229960001967 tacrolimus Drugs 0.000 description 11
- QJJXYPPXXYFBGM-SHYZHZOCSA-N tacrolimus Natural products CO[C@H]1C[C@H](CC[C@@H]1O)C=C(C)[C@H]2OC(=O)[C@H]3CCCCN3C(=O)C(=O)[C@@]4(O)O[C@@H]([C@H](C[C@H]4C)OC)[C@@H](C[C@H](C)CC(=C[C@@H](CC=C)C(=O)C[C@H](O)[C@H]2C)C)OC QJJXYPPXXYFBGM-SHYZHZOCSA-N 0.000 description 11
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 10
- 102000004127 Cytokines Human genes 0.000 description 10
- 108090000695 Cytokines Proteins 0.000 description 10
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 10
- 125000003275 alpha amino acid group Chemical group 0.000 description 10
- 230000008859 change Effects 0.000 description 10
- 239000012636 effector Substances 0.000 description 10
- 229960001008 heparin sodium Drugs 0.000 description 10
- 102100028990 C-X-C chemokine receptor type 3 Human genes 0.000 description 9
- 101000916050 Homo sapiens C-X-C chemokine receptor type 3 Proteins 0.000 description 9
- 102100025323 Integrin alpha-1 Human genes 0.000 description 9
- 241001465754 Metazoa Species 0.000 description 9
- 210000001744 T-lymphocyte Anatomy 0.000 description 9
- 238000005138 cryopreservation Methods 0.000 description 9
- 239000003814 drug Substances 0.000 description 9
- 238000002360 preparation method Methods 0.000 description 9
- 102100035875 C-C chemokine receptor type 5 Human genes 0.000 description 8
- 101710149870 C-C chemokine receptor type 5 Proteins 0.000 description 8
- 102100025074 C-C chemokine receptor-like 2 Human genes 0.000 description 8
- 101000716068 Homo sapiens C-C chemokine receptor type 6 Proteins 0.000 description 8
- 101001078158 Homo sapiens Integrin alpha-1 Proteins 0.000 description 8
- 101000994378 Homo sapiens Integrin alpha-3 Proteins 0.000 description 8
- 102100032819 Integrin alpha-3 Human genes 0.000 description 8
- 230000003321 amplification Effects 0.000 description 8
- 230000000259 anti-tumor effect Effects 0.000 description 8
- 230000000694 effects Effects 0.000 description 8
- 238000003199 nucleic acid amplification method Methods 0.000 description 8
- 201000011510 cancer Diseases 0.000 description 7
- 238000005259 measurement Methods 0.000 description 7
- 101001023379 Homo sapiens Lysosome-associated membrane glycoprotein 1 Proteins 0.000 description 6
- 108090000171 Interleukin-18 Proteins 0.000 description 6
- 102100035133 Lysosome-associated membrane glycoprotein 1 Human genes 0.000 description 6
- 210000000601 blood cell Anatomy 0.000 description 6
- 239000001963 growth medium Substances 0.000 description 6
- 210000001616 monocyte Anatomy 0.000 description 6
- 102000004169 proteins and genes Human genes 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 5
- 229960004669 basiliximab Drugs 0.000 description 5
- 230000004663 cell proliferation Effects 0.000 description 5
- 239000002577 cryoprotective agent Substances 0.000 description 5
- 238000005516 engineering process Methods 0.000 description 5
- 239000011259 mixed solution Substances 0.000 description 5
- 239000013642 negative control Substances 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 229960005322 streptomycin Drugs 0.000 description 5
- 238000010257 thawing Methods 0.000 description 5
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 4
- 108091006905 Human Serum Albumin Proteins 0.000 description 4
- 102000008100 Human Serum Albumin Human genes 0.000 description 4
- 102000015696 Interleukins Human genes 0.000 description 4
- 108010063738 Interleukins Proteins 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- 206010033128 Ovarian cancer Diseases 0.000 description 4
- 206010061535 Ovarian neoplasm Diseases 0.000 description 4
- 230000009089 cytolysis Effects 0.000 description 4
- 230000003013 cytotoxicity Effects 0.000 description 4
- 231100000135 cytotoxicity Toxicity 0.000 description 4
- 229940079593 drug Drugs 0.000 description 4
- 229960002897 heparin Drugs 0.000 description 4
- OOYGSFOGFJDDHP-KMCOLRRFSA-N kanamycin A sulfate Chemical compound OS(O)(=O)=O.O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N OOYGSFOGFJDDHP-KMCOLRRFSA-N 0.000 description 4
- 229960002064 kanamycin sulfate Drugs 0.000 description 4
- 208000032839 leukemia Diseases 0.000 description 4
- 210000004698 lymphocyte Anatomy 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- 210000000130 stem cell Anatomy 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- 239000000725 suspension Substances 0.000 description 4
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 3
- 206010002091 Anaesthesia Diseases 0.000 description 3
- 101000638251 Homo sapiens Tumor necrosis factor ligand superfamily member 9 Proteins 0.000 description 3
- 108090001007 Interleukin-8 Proteins 0.000 description 3
- 108060001084 Luciferase Proteins 0.000 description 3
- 239000005089 Luciferase Substances 0.000 description 3
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 3
- 230000037005 anaesthesia Effects 0.000 description 3
- 239000003242 anti bacterial agent Substances 0.000 description 3
- 210000000612 antigen-presenting cell Anatomy 0.000 description 3
- 239000011324 bead Substances 0.000 description 3
- 230000003115 biocidal effect Effects 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- 230000010261 cell growth Effects 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 201000010099 disease Diseases 0.000 description 3
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 3
- 210000001671 embryonic stem cell Anatomy 0.000 description 3
- 229960003444 immunosuppressant agent Drugs 0.000 description 3
- 239000003018 immunosuppressive agent Substances 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 238000011503 in vivo imaging Methods 0.000 description 3
- 238000007912 intraperitoneal administration Methods 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- 239000013641 positive control Substances 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 238000003908 quality control method Methods 0.000 description 3
- 230000006798 recombination Effects 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 238000002054 transplantation Methods 0.000 description 3
- 238000011282 treatment Methods 0.000 description 3
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 3
- YXHLJMWYDTXDHS-IRFLANFNSA-N 7-aminoactinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=C(N)C=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 YXHLJMWYDTXDHS-IRFLANFNSA-N 0.000 description 2
- 108700012813 7-aminoactinomycin D Proteins 0.000 description 2
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 2
- 206010009944 Colon cancer Diseases 0.000 description 2
- 208000035473 Communicable disease Diseases 0.000 description 2
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 2
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 2
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 2
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 2
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 2
- 102100037850 Interferon gamma Human genes 0.000 description 2
- 108010074328 Interferon-gamma Proteins 0.000 description 2
- 108010050904 Interferons Proteins 0.000 description 2
- 102000014150 Interferons Human genes 0.000 description 2
- 102000003812 Interleukin-15 Human genes 0.000 description 2
- 108090000172 Interleukin-15 Proteins 0.000 description 2
- PIWKPBJCKXDKJR-UHFFFAOYSA-N Isoflurane Chemical compound FC(F)OC(Cl)C(F)(F)F PIWKPBJCKXDKJR-UHFFFAOYSA-N 0.000 description 2
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 2
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 2
- 108010004222 Natural Cytotoxicity Triggering Receptor 3 Proteins 0.000 description 2
- 102100032852 Natural cytotoxicity triggering receptor 3 Human genes 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- 230000000996 additive effect Effects 0.000 description 2
- 229940125644 antibody drug Drugs 0.000 description 2
- 230000000890 antigenic effect Effects 0.000 description 2
- 238000002617 apheresis Methods 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 210000000988 bone and bone Anatomy 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 238000002659 cell therapy Methods 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 208000029742 colonic neoplasm Diseases 0.000 description 2
- 238000012136 culture method Methods 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 108700014844 flt3 ligand Proteins 0.000 description 2
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 2
- 239000012737 fresh medium Substances 0.000 description 2
- 238000011194 good manufacturing practice Methods 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 230000001861 immunosuppressant effect Effects 0.000 description 2
- 210000004263 induced pluripotent stem cell Anatomy 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 102000006495 integrins Human genes 0.000 description 2
- 108010044426 integrins Proteins 0.000 description 2
- 229940079322 interferon Drugs 0.000 description 2
- 229960002725 isoflurane Drugs 0.000 description 2
- 238000004020 luminiscence type Methods 0.000 description 2
- 239000003002 pH adjusting agent Substances 0.000 description 2
- 229950001699 teceleukin Drugs 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 229960000575 trastuzumab Drugs 0.000 description 2
- PNDPGZBMCMUPRI-HVTJNCQCSA-N 10043-66-0 Chemical compound [131I][131I] PNDPGZBMCMUPRI-HVTJNCQCSA-N 0.000 description 1
- RTQWWZBSTRGEAV-PKHIMPSTSA-N 2-[[(2s)-2-[bis(carboxymethyl)amino]-3-[4-(methylcarbamoylamino)phenyl]propyl]-[2-[bis(carboxymethyl)amino]propyl]amino]acetic acid Chemical compound CNC(=O)NC1=CC=C(C[C@@H](CN(CC(C)N(CC(O)=O)CC(O)=O)CC(O)=O)N(CC(O)=O)CC(O)=O)C=C1 RTQWWZBSTRGEAV-PKHIMPSTSA-N 0.000 description 1
- UZOVYGYOLBIAJR-UHFFFAOYSA-N 4-isocyanato-4'-methyldiphenylmethane Chemical compound C1=CC(C)=CC=C1CC1=CC=C(N=C=O)C=C1 UZOVYGYOLBIAJR-UHFFFAOYSA-N 0.000 description 1
- MJZJYWCQPMNPRM-UHFFFAOYSA-N 6,6-dimethyl-1-[3-(2,4,5-trichlorophenoxy)propoxy]-1,6-dihydro-1,3,5-triazine-2,4-diamine Chemical compound CC1(C)N=C(N)N=C(N)N1OCCCOC1=CC(Cl)=C(Cl)C=C1Cl MJZJYWCQPMNPRM-UHFFFAOYSA-N 0.000 description 1
- 239000012099 Alexa Fluor family Substances 0.000 description 1
- 208000025324 B-cell acute lymphoblastic leukemia Diseases 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 206010004593 Bile duct cancer Diseases 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 1
- 229940122739 Calcineurin inhibitor Drugs 0.000 description 1
- 101710192106 Calcineurin-binding protein cabin-1 Proteins 0.000 description 1
- 102100024123 Calcineurin-binding protein cabin-1 Human genes 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 101000957724 Catostomus commersonii Corticoliberin-1 Proteins 0.000 description 1
- 102000009410 Chemokine receptor Human genes 0.000 description 1
- 108050000299 Chemokine receptor Proteins 0.000 description 1
- 241000725101 Clea Species 0.000 description 1
- 235000005956 Cosmos caudatus Nutrition 0.000 description 1
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 1
- 108010036949 Cyclosporine Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 208000022072 Gallbladder Neoplasms Diseases 0.000 description 1
- 208000032612 Glial tumor Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 1
- 101000976075 Homo sapiens Insulin Proteins 0.000 description 1
- 101001015004 Homo sapiens Integrin beta-3 Proteins 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 1
- 101000766306 Homo sapiens Serotransferrin Proteins 0.000 description 1
- 108010041341 Integrin alpha1 Proteins 0.000 description 1
- 108010041357 Integrin alpha3 Proteins 0.000 description 1
- 102000000510 Integrin alpha3 Human genes 0.000 description 1
- 102100032999 Integrin beta-3 Human genes 0.000 description 1
- 108010065805 Interleukin-12 Proteins 0.000 description 1
- 108010002386 Interleukin-3 Proteins 0.000 description 1
- 108010002586 Interleukin-7 Proteins 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 1
- 208000003445 Mouth Neoplasms Diseases 0.000 description 1
- 208000010428 Muscle Weakness Diseases 0.000 description 1
- 206010028372 Muscular weakness Diseases 0.000 description 1
- 102000004473 OX40 Ligand Human genes 0.000 description 1
- 108010042215 OX40 Ligand Proteins 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 229960000446 abciximab Drugs 0.000 description 1
- 210000000683 abdominal cavity Anatomy 0.000 description 1
- 229960002964 adalimumab Drugs 0.000 description 1
- 238000004378 air conditioning Methods 0.000 description 1
- 229960000548 alemtuzumab Drugs 0.000 description 1
- 229960004539 alirocumab Drugs 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 238000003975 animal breeding Methods 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 210000001367 artery Anatomy 0.000 description 1
- 239000007640 basal medium Substances 0.000 description 1
- 229960003270 belimumab Drugs 0.000 description 1
- 229960000397 bevacizumab Drugs 0.000 description 1
- 208000026900 bile duct neoplasm Diseases 0.000 description 1
- 229960003008 blinatumomab Drugs 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 229960000455 brentuximab vedotin Drugs 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 229960001838 canakinumab Drugs 0.000 description 1
- 238000002619 cancer immunotherapy Methods 0.000 description 1
- 239000012159 carrier gas Substances 0.000 description 1
- 229960000419 catumaxomab Drugs 0.000 description 1
- 230000021164 cell adhesion Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 229960005395 cetuximab Drugs 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 208000006990 cholangiocarcinoma Diseases 0.000 description 1
- 229960001265 ciclosporin Drugs 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000000139 costimulatory effect Effects 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 230000001186 cumulative effect Effects 0.000 description 1
- 229930182912 cyclosporin Natural products 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 229960002806 daclizumab Drugs 0.000 description 1
- 229960002204 daratumumab Drugs 0.000 description 1
- 229960001251 denosumab Drugs 0.000 description 1
- 238000000432 density-gradient centrifugation Methods 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 229960001259 diclofenac Drugs 0.000 description 1
- DCOPUUMXTXDBNB-UHFFFAOYSA-N diclofenac Chemical compound OC(=O)CC1=CC=CC=C1NC1=C(Cl)C=CC=C1Cl DCOPUUMXTXDBNB-UHFFFAOYSA-N 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 230000035622 drinking Effects 0.000 description 1
- 229960002224 eculizumab Drugs 0.000 description 1
- 229960000284 efalizumab Drugs 0.000 description 1
- 229960004137 elotuzumab Drugs 0.000 description 1
- 229960002027 evolocumab Drugs 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 210000004700 fetal blood Anatomy 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- -1 for example Chemical compound 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 201000010175 gallbladder cancer Diseases 0.000 description 1
- 230000005251 gamma ray Effects 0.000 description 1
- 229960000578 gemtuzumab Drugs 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 229960001743 golimumab Drugs 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 102000055277 human IL2 Human genes 0.000 description 1
- 229960001001 ibritumomab tiuxetan Drugs 0.000 description 1
- 229960002308 idarucizumab Drugs 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 229960000598 infliximab Drugs 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 230000015788 innate immune response Effects 0.000 description 1
- PBGKTOXHQIOBKM-FHFVDXKLSA-N insulin (human) Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 PBGKTOXHQIOBKM-FHFVDXKLSA-N 0.000 description 1
- 229940047122 interleukins Drugs 0.000 description 1
- 230000009878 intermolecular interaction Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- 229960005435 ixekizumab Drugs 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 208000012987 lip and oral cavity carcinoma Diseases 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 201000007270 liver cancer Diseases 0.000 description 1
- 208000014018 liver neoplasm Diseases 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- FVVLHONNBARESJ-NTOWJWGLSA-H magnesium;potassium;trisodium;(2r,3s,4r,5r)-2,3,4,5,6-pentahydroxyhexanoate;acetate;tetrachloride;nonahydrate Chemical compound O.O.O.O.O.O.O.O.O.[Na+].[Na+].[Na+].[Mg+2].[Cl-].[Cl-].[Cl-].[Cl-].[K+].CC([O-])=O.OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O FVVLHONNBARESJ-NTOWJWGLSA-H 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 229960005108 mepolizumab Drugs 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 239000007758 minimum essential medium Substances 0.000 description 1
- 229950007699 mogamulizumab Drugs 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 229960003816 muromonab-cd3 Drugs 0.000 description 1
- 229960005027 natalizumab Drugs 0.000 description 1
- 229960000513 necitumumab Drugs 0.000 description 1
- 229960003301 nivolumab Drugs 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 229960003419 obiltoxaximab Drugs 0.000 description 1
- 229960003347 obinutuzumab Drugs 0.000 description 1
- 229960002450 ofatumumab Drugs 0.000 description 1
- 229960000470 omalizumab Drugs 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 229960000402 palivizumab Drugs 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229960002087 pertuzumab Drugs 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- XNSAINXGIQZQOO-SRVKXCTJSA-N protirelin Chemical compound NC(=O)[C@@H]1CCCN1C(=O)[C@@H](NC(=O)[C@H]1NC(=O)CC1)CC1=CN=CN1 XNSAINXGIQZQOO-SRVKXCTJSA-N 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 239000000941 radioactive substance Substances 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 229960002633 ramucirumab Drugs 0.000 description 1
- 229960003876 ranibizumab Drugs 0.000 description 1
- 229960004910 raxibacumab Drugs 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000001172 regenerating effect Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 229960003254 reslizumab Drugs 0.000 description 1
- 229960004641 rituximab Drugs 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 229960004540 secukinumab Drugs 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 229960003323 siltuximab Drugs 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 239000008399 tap water Substances 0.000 description 1
- 235000020679 tap water Nutrition 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 229960003989 tocilizumab Drugs 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 102000027257 transmembrane receptors Human genes 0.000 description 1
- 108091008578 transmembrane receptors Proteins 0.000 description 1
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 229960003824 ustekinumab Drugs 0.000 description 1
- 229950003520 utomilumab Drugs 0.000 description 1
- 229960004914 vedolizumab Drugs 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N1/00—Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
Definitions
- the present invention relates to a method for producing or amplifying and activating a cell population containing CD3-negative and CD56-positive cells.
- Natural killer cells are a type of cytotoxic lymphocyte that have a surface marker phenotype of CD3 negative and CD56 positive, and act as a key factor in innate immunity.
- Non-Patent Document 1 Methods for producing cell populations that exhibit high cytotoxicity against solid tumors are known (Patent Document 1, Non-Patent Document 2).
- Non-Patent Document 3 describes NK cells derived from human iPS cells that express a CAR containing the transmembrane domain of NK2GD, the 2B4 (CD244) costimulatory domain, and the CD3 ⁇ signal domain.
- Patent Document 4 describes a method for expanding tumor-infiltrating lymphocytes using tumor necrosis factor receptor superfamily agonists such as 4-1BB agonists.
- Patent Document 3 describes a liquid composition for expanding T cells, which contains IL-2, IL-15, and IL-21 at predetermined concentrations.
- Non-Patent Document 4 describes that under the experimental conditions, when 4-1BBL or IL-21 is used as a humoral factor, the NK cell expansion effect is inferior compared to when these are immobilized on beads, suggesting that using factors such as 4-1BBL and IL-21 as humoral factors is inefficient. Therefore, a means for realizing high NK cell proliferation efficiency by using a genetically modified feeder cell line or a synthetic lipid membrane has been developed.
- Non-Patent Document 5 describes a method for co-culturing NK cells with genetically modified artificial antigen presenting cells expressing membrane-bound IL-21.
- Patent Document 4 describes a method for producing NK cells, in which peripheral blood mononuclear cells from which CD3 positive cells have been removed are grown together with feeder cells, and when the peripheral blood mononuclear cells reach a specific cumulative division level, the cells are restimulated with feeder cells.
- Patent Publication 2019-170176 Special table 2020-514289 Patent Publication No. 2021-113200 Special table 2021-518122
- CAR-NK Chimeric antigen receptor natural killer
- Non-Patent Document 3 In preparations using NK cells derived from ES cells/iPS cells such as those in Non-Patent Document 3, activity against solid tumors is particularly insufficient. Furthermore, the inventions described in Patent Documents 2 and 3 relate to the proliferation of T cells, and have not been shown to be useful for the proliferation of NK cells. In methods using gene recombination techniques or feeder cell lines such as those in Non-Patent Document 5 and Patent Document 4, the hurdles of quality control are increased, the manufacturing/purification processes become complicated (Non-Patent Document 6), and the burden of medical expenses may also increase.
- the present invention aims to efficiently grow a cell population containing CD3-negative and CD56-positive cells without using a feeder cell line or genetic recombination techniques.
- the inventors conducted extensive research to solve the above problems and discovered that a method including the steps of: a) culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a medium containing IL-2, IL-21 and a 4-1BB agonist, where the IL-21 and the 4-1BB agonist are humoral factors, can mass-produce and amplify a cell population containing CD3-negative and CD56-positive cells, as compared to conventional manufacturing methods, and thus completed the present invention.
- a method for producing a cell population containing CD3-negative and CD56-positive cells comprising the steps of: a) culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a medium containing IL-2, IL-21 and a 4-1BB agonist for one day or more, wherein the IL-21 and the 4-1BB agonist are humoral factors.
- [2] The method according to [1], further comprising the steps of: b) Passaging the cell population cultured in step a) in a medium containing IL-2 and a 4-1BB agonist but not containing IL-21, and culturing the medium for one day or more.
- [3] The method for production according to [1] or [2], wherein the concentration of the 4-1BB agonist in the medium in step a) is 1 ⁇ g/mL or more, and the concentration of IL-21 in the medium in step a) is 1 ng/mL or more.
- [4] The method according to [2], further comprising the steps of: c) Passaging the cell population cultured in step b) in a medium containing IL-2 and culturing for one day or more.
- the 4-1BB agonist is selected from an anti-4-1BB antibody and a 4-1BB ligand.
- the medium contains a platelet lysate.
- a cell population containing CD3-negative and CD56-positive cells can be mass-multiplied and obtained without using a feeder cell line or genetic recombination techniques, as compared to conventional manufacturing methods.
- FIG. 1 shows the results (changes in cell number) of a NK cell culture test carried out under different conditions of culture medium and additives.
- FIG. 2 shows the results of a culture test in which the conditions for the cells used as the culture material were changed and compared (changes in the amplification fold of the cells).
- FIG. 3 shows the results of flow cytometry performed for each cell surface marker.
- FIG. 4 shows the results (changes in cell number) of a culture test comparing different concentrations of anti-4-1BB antibody and IL-21.
- FIG. 5 shows the results of a culture test comparing different concentrations of anti-4-1BB antibody and IL-21 (changes in cell amplification fold).
- FIG. 6 shows the results of flow cytometry performed for each cell surface marker.
- FIG. 1 shows the results (changes in cell number) of a NK cell culture test carried out under different conditions of culture medium and additives.
- FIG. 2 shows the results of a culture test in which the conditions for the cells used as the culture material were changed and
- FIG. 7 shows the results of flow cytometry for CD3/CD56 or CXCR3 performed using the Day 14 cell population, and the results of measuring the cytotoxic activity against K562 cells using the Day 17 cell population.
- FIG. 8 shows the results of examining the antitumor effect of administration of the recovered cell population using cancer-bearing mice generated using SK-OV-3/CMV-Luc (a luciferase-expressing human ovarian cancer cell line) (changes in the number of SK-OV-3/CMV-Luc cells).
- Figure 9 shows the results of verifying the antitumor effect of administration of the recovered cell population using tumor-bearing mice generated using SK-OV-3/CMV-Luc (IVIS (in vivo imaging system) results on Day 69).
- FIG. 10 shows the results of flow cytometry for CD3/CD56 or CXCR4.
- FIG. 11 shows the results of measuring the cytotoxic activity against K562 cells and Raji cells using the recovered cell population.
- FIG. 12 shows the results of a culture test (changes in cell amplification fold) when Simulect and tacrolimus were added to the medium.
- FIG. 13 shows the results of a culture test (changes in cell amplification fold) when recombinant 4-1BB ligand was used instead of anti-4-1BB antibody.
- FIG. 14 shows the results of flow cytometry performed for each cell surface marker.
- FIG. 15 shows the results of flow cytometry performed for each cell surface marker.
- a method for producing a cell population containing CD3-negative and CD56-positive cells comprising the steps of: a) a step of culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a predetermined medium.
- a cell population refers to a group of cells consisting of a plurality of cells, for example, 1 ⁇ 10 5 cells or more.
- the cell population provided by the present invention can be prepared to various cell densities. For example, it can be prepared to 1 ⁇ 10 5 cells/mL or more.
- a cell population containing at least CD3-negative and CD56-positive cells is obtained. Whether or not a cell is CD3-negative and CD56-positive can be determined by analyzing the expression pattern of the cell surface marker. In the present invention, whether or not a cell surface marker is positive can be determined, for example, by flow cytometry, by comparing the signal from the cell surface marker with that of a control cell population that does not express or expresses the target cell surface marker at a low level. Whether or not a cell surface marker is negative can also be determined by a similar method.
- a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing monocytes can be used.
- the cell population containing monocytes can contain various lymphocytes such as T cells, B cells, NK cells, monocytes, and dendritic cells.
- the cell population containing monocytes can be peripheral blood mononuclear cells obtained by separating monocytes from blood cells collected from a donor. Blood cells can be collected from a donor by a method known to those skilled in the art, for example, by apheresis from the donor's peripheral blood. Blood cells may be collected from the donor's umbilical cord blood, bone marrow, lymph nodes, etc.
- the separation of mononuclear cells from blood cells can be performed, for example, by density gradient centrifugation.
- Blood cells obtained by differentiating embryonic stem cells (ES cells) or induced pluripotent stem cells (iPS cells) can also be used.
- the method for differentiating into blood cells can be performed by a method known to those skilled in the art, for example, by apheresis from the donor's peripheral blood.
- the methods described in Melichar et al. Comparative Study of Hematopoietic Differentiation between Human Embryonic Stem Cell Lines. PLoS One. 2011; 6(5): e19854. doi: 10.1371/journal. pone. 0019854. and the like can be referred to.
- Removal of CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells may be performed according to a method known to those skilled in the art, for example, by a method of separating CD3-positive cells or CD34-positive cells using magnetic beads bound to an antibody against CD3 or CD34.
- the cell population containing mononuclear cells may be derived from a single donor or from multiple donors.
- the proportion of the cell population derived from each donor in the cell population is not particularly limited.
- the cell population containing mononuclear cells may be used after being thawed from cryopreservation.
- Cryopreservation can be performed, for example, by suspending the cell population containing mononuclear cells to an appropriate cell density in a medium suitable for cryopreservation, such as a complete medium containing a cryoprotective agent such as dimethyl sulfoxide (DMSO), placing it in a cryopreservation container such as a cryovial, and storing it at -80°C or below.
- Thawing can be performed, for example, by transferring the cryopreservation container containing the cell population containing mononuclear cells to a 37°C water bath and gently swirling it.
- the method for producing a cell population of the present invention includes a step a) of culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a predetermined medium.
- the medium may further contain additives, reagents, etc. suitable for culture.
- “medium in the method for producing a cell population of the present invention” “medium of the present invention”, “medium” or the like, and not particularly specifying "medium in step a)” or “medium in step b"), etc., it refers to the medium in one or more selected from step a), step b) and step c).
- the concentration refers to the concentration in the medium at the start of each step, such as step a), step b), step c), etc., and does not specify the concentration throughout the entire period of each step.
- the medium in the method for producing a cell population of the present invention may contain a cytokine, polypeptide, etc. suitable for expanding the cell population.
- Suitable cytokines are, for example, one or more interleukins (IL), preferably IL-2 and IL-21.
- Suitable polypeptides are preferably 4-1BB agonists.
- 4-1BB belongs to the tumor necrosis factor receptor superfamily (TNFRSF) and is a transmembrane receptor that is mainly expressed on the cell surface of lymphocytes. 4-1BB is also called CD137, TNFRSF9, etc.
- a 4-1BB agonist refers to one that can bind to 4-1BB and promote its function.
- 4-1BB examples include the production of IL-8 from human peripheral blood mononuclear cells and the production of interferon (IFN)- ⁇ from human T lymphocytes, and the production of IL-8 and IFN- ⁇ by 4-1BB can be measured by a method known to those skilled in the art.
- the 4-1BB agonist may be, for example, one that can bind to 4-1BB and promote the production of IL-8 or IFN- ⁇ .
- Examples of the 4-1BB agonist include anti-4-1BB antibodies and 4-1BB ligand (4-1BBL).
- anti-4-1BB antibodies refer to antibodies that can recognize and bind to one or more specific antigenic determinants in 4-1BB and can multimerize 4-1BB, such as urelumab, utomilumab, or antibodies that can recognize the same antigenic determinants as these.
- 4-1BB ligand refers to a polypeptide or protein that can bind to 4-1BB (excluding antibodies).
- 4-1BBL may be synthesized by a method known to those skilled in the art, or may be artificially produced by recombinant gene technology, and 4-1BBL artificially produced by recombinant gene technology is called recombinant 4-1BBL.
- 4-1BBL may be a polypeptide consisting of the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2 shown below, a polypeptide containing the extracellular domain of 4-1BBL (SEQ ID NO: 3), or a polypeptide consisting of an amino acid sequence having at least 70% or more, 80% or more, 85% or more, 90% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more identity with SEQ ID NO: 1, SEQ ID NO: 2, or SEQ ID NO: 3, and having proliferation activity of CD3-negative and CD56-positive cells.
- Amino acid sequence identity refers to the identity of the amino acid sequences between the target polypeptides, and is expressed as the percentage (%) of amino acid residues that match in the optimal alignment of the amino acid sequences. Amino acid sequence identity can be calculated using, for example, a homology search program known to those skilled in the art, such as BLAST or FASTA.
- a polypeptide having proliferation activity of CD3-negative and CD56-positive cells refers to a polypeptide that, when used in the cell culture method described in the Examples of this specification, can amplify the number of cells in a cell population containing CD3-negative and CD56-positive cells by at least two-fold, preferably three-fold.
- the 4-1BB agonist of the present invention is selected from the group consisting of urelumab, utomirumab, and a 4-1BB ligand.
- the 4-1BB agonist is one or more selected from urelumab and a polypeptide having an amino acid sequence with 90% or more identity to SEQ ID NO: 1 and having proliferation activity of CD3-negative and CD56-positive cells. It is preferable that the cytokine, polypeptide, etc. used have a human amino acid sequence.
- the medium in step a) of the method for producing a cell population of the present invention contains IL-2, IL-21 and a 4-1BB agonist.
- the concentration of IL-2 in the medium of the present invention is not particularly limited, and can be 50 IU/mL or more, preferably 100 IU/mL or more, more preferably 150 IU/mL or more, even more preferably 200 IU/mL or more, particularly preferably 250 IU/mL or more, and most preferably 281 IU/mL or more.
- the concentration of IL-2 in the medium in step a) may be, for example, 3,000 IU/mL or less, 2,810 IU/mL or less, 2,500 IU/mL or less, 2,000 IU/mL or less, 1,500 IU/mL or less, 1,000 IU/mL or less, 500 IU/mL or less, or 300 IU/mL or less.
- the upper and lower limits of any of the above-mentioned IL-2 concentrations may be combined to represent the concentration range of IL-2.
- the concentration of IL-21 in the medium in step a) of the production method of the present invention is not particularly limited, and may be 0.01 ng/mL or more, preferably 0.05 ng/mL or more, more preferably 0.1 ng/mL or more, even more preferably 0.5 ng/mL or more, and particularly preferably 1 ng/mL or more.
- the concentration of IL-21 in the medium in step a) may be, for example, 1000 ng/mL or less, may be 500 ng/mL or less, preferably 100 ng/mL or less, more preferably 50 ng/mL or less, even more preferably 10 ng/mL or less, and may be 9 ng/mL or less, 8 ng/mL or less, 7 ng/mL or less, 6 ng/mL or less, 5 ng/mL or less, 4 ng/mL or less, 3 ng/mL or less, or 2 ng/mL or less.
- the upper and lower limits of any of the above-mentioned IL-21 concentrations may be combined to represent the concentration range of IL-21.
- the concentration of the 4-1BB agonist in the medium in step a) of the production method of the present invention is not particularly limited, and can be 0.01 ⁇ g/mL or more, preferably 0.05 ⁇ g/mL or more, more preferably 0.1 ⁇ g/mL or more, even more preferably 0.5 ⁇ g/mL or more, and particularly preferably 1 ⁇ g/mL or more.
- the concentration of the 4-1BB agonist in the medium in step a) is not particularly limited, and can be, for example, 1000 ⁇ g/mL or less, preferably 500 ⁇ g/mL or less, more preferably 100 ⁇ g/mL or less, and even more preferably 50 ⁇ g/mL or less.
- the upper and lower limits of any of the above-mentioned concentrations of the 4-1BB agonist may be combined to express the concentration range of the 4-1BB agonist.
- the concentration of the anti-4-1BB antibody in the medium in step a) of the production method of the present invention can be 0.01 ⁇ g/mL or more, preferably 0.05 ⁇ g/mL or more, more preferably 0.1 ⁇ g/mL or more, even more preferably 0.5 ⁇ g/mL or more, and particularly preferably 1 ⁇ g/mL or more.
- the concentration of 4-1BBL in the medium in step a) is preferably 100 ⁇ g/mL or less, more preferably 50 ⁇ g/mL or less, even more preferably 10 ⁇ g/mL or less, and may be 9 ⁇ g/mL or less, 8 ⁇ g/mL or less, 7 ⁇ g/mL or less, 6 ⁇ g/mL or less, 5 ⁇ g/mL or less, 4 ⁇ g/mL or less, 3 ⁇ g/mL or less, or 2 ⁇ g/mL or less.
- the concentration of anti-4-1BBL in the medium in step a) of the production method of the present invention can be 0.1 ⁇ g/mL or more, preferably 0.5 ⁇ g/mL or more, more preferably 1 ⁇ g/mL or more, even more preferably 2 ⁇ g/mL or more, 3 ⁇ g/mL or more, 4 ⁇ g/mL or more, or 5 ⁇ g/mL or more, and particularly preferably 10 ⁇ g/mL or more.
- the concentration of 4-1BBL in the medium in step a) can be, for example, 1000 ⁇ g/mL or less, preferably 500 ⁇ g/mL or less, more preferably 100 ⁇ g/mL or less, and even more preferably 50 ⁇ g/mL or less.
- IL-21 and 4-1BB agonists are humoral factors.
- "Hydrologic" means that the target protein, cytokine, or other substance is not immobilized and can dissolve in the medium, and humoral factors refer to proteins, etc. that are liquid.
- Opposite concepts include, for example, a state in which the target protein, etc. is immobilized on beads as described in Non-Patent Document 4, and a state in which the target protein, etc. is expressed on the cell membrane of the feeder cells after transformation as described in Non-Patent Document 5.
- the medium containing humoral factors is preferably liquid.
- the medium of the present invention is not particularly limited, and examples thereof include KBM501 medium (KOHJIN BIO), KBM502 medium (KOHJIN BIO), NTI medium (FUKOKU), Cosmedium 008 (Cosmo Bio), FKCM101 (FUKOKU), CellGro SCGM medium (CellGenics, Iwai Chemicals Co., Ltd.), X-VIVO15 medium (Lonza, Takara Bio Inc.), Gibco (registered trademark) CTS (registered trademark) AIM V (registered trademark) Medium (Thermo Fisher Scientific, a chemically defined serum-free medium for growing and manipulating T cells and dendritic cells), CTS OpTmizer T Cell Expansion Basal Medium (Thermo Fisher Scientific, for growth and proliferation of human T lymphocytes), IMDM, MEM, DMEM, RPMI-1640.
- KBM501 medium KOHJIN BIO
- KBM502 medium KOHJIN BIO
- NTI medium FUKOKU
- the term “culturing” cells refers to maintaining cells in a medium or a liquid equivalent thereto for a certain period of time for any purpose selected from the group consisting of maintaining cell survival, expanding cells, and activating cells. "Incubating” may be used when a process is carried out at a specific temperature for a certain period of time.
- the medium may contain IL-2.
- media containing IL-2 include KBM501 medium (containing 2,810 IU/mL (1,750 JRU/mL) of IL-2), KBM502 medium (containing 281 IU/mL (175 JRU/mL) of IL-2), Cosmedium 008 medium (containing 1,750 JRU/mL of IL-2), FKCM101-L300 medium (FUKOKU, containing 300 IU/mL of IL-2), and FKCM101-L13T medium (FUKOKU, containing 1,300 IU/mL of IL-2).
- the medium may contain KBM502 medium or KBM501 medium.
- the medium may be replaced or replenished at any time after the start of culture, provided that the desired culture effect is obtained, but it is preferable to do so every 2 to 5 days.
- the culture vessels used for the culture include, but are not limited to, commercially available dishes, flasks, plates, and multi-well plates. In addition to the culture vessels, various culture materials such as supports may be used for the culture.
- the culture conditions are not particularly limited as long as they do not impair the culture effect, but the culture conditions are generally 37°C, 5% CO2, and saturated water vapor atmosphere.
- the concentration of basiliximab in the medium may be, for example, 0.005 to 50 ⁇ g/mL, preferably 0.05 to 30 ⁇ g/mL, and more preferably 0.5 to 10 ⁇ g/mL.
- tacrolimus for example, Prograf injection available from Astellas Pharma can be used.
- Basiliximab can be, for example, Simulect for intravenous injection, available from Novartis Pharma.
- Step a) may include culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells for a predetermined period of time.
- the predetermined period may be 1 day or more, or 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, 11 days or more, 12 days or more, 13 days or more, 14 days or more, or more, and may be 30 days or less, 25 days or less, 20 days or less, 15 days or less, or less, and is not particularly limited.
- passaging refers to transferring a cell population to a new medium
- the method is not particularly limited. Transferring a cell population to a new medium may involve, for example, collecting a portion of the medium in which the cells are dispersed and adding it to the new medium.
- passaging a portion of the medium used for the culture before passaging may be added to the new medium, but in this specification, the composition of the medium used for the culture before passaging can be ignored in the composition of the new medium after passaging.
- the concentration of IL-21 in the medium may be, for example, 0.01 ng/mL or more, 0.1 ng/mL or more, or 1 ng/mL or more, while it is preferably 100 ng/mL or less, more preferably 50 ng/mL or less, even more preferably 10 ng/mL or less, and may be 9 ⁇ g/mL or less, 8 ⁇ g/mL or less, 7 ⁇ g/mL or less, 6 ⁇ g/mL or less, 5 ⁇ g/mL or less, 4 ⁇ g/mL or less, 3 ⁇ g/mL or less, or 2 ⁇ g/mL or less.
- the concentration of the anti-4-1BB antibody in the medium in step b) can be 0.01 ⁇ g/mL or more, preferably 0.05 ⁇ g/mL or more, more preferably 0.1 ⁇ g/mL or more, even more preferably 0.5 ⁇ g/mL or more, and particularly preferably 1 ⁇ g/mL or more.
- the concentration of the anti-4-1BB antibody in the medium in step b) is preferably equal to or higher than the concentration of the anti-4-1BB antibody in the medium in step a).
- the concentration of 4-1BBL in the medium in step b) can be 0.1 ⁇ g/mL or more, preferably 0.5 ⁇ g/mL or more, more preferably 1 ⁇ g/mL or more, even more preferably 5 ⁇ g/mL or more, and particularly preferably 10 ⁇ g/mL or more.
- the concentration of 4-1BBL in the medium in step b) is preferably equal to or greater than the concentration of 4-1BBL in the medium in step a).
- Step b) may include culturing the cell population cultured in step a) for a predetermined period of time.
- the predetermined period may be 1 day or more, or 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, 11 days or more, 12 days or more, 13 days or more, 14 days or more, 15 days or more, or more, and may be 30 days or less, 25 days or less, 20 days or less, or less, and is not particularly limited.
- the culture period in step b) may be, for example, 1 to 2 days, preferably 2 to 21 days, more preferably 3 to 19 days, even more preferably 4 to 17 days, and particularly preferably 8 to 15 days.
- the cells may be added to the same medium, or the medium may be replaced (in this case, it can be said that step b) is continued as long as the medium meets the requirements).
- the method for producing a cell population according to the present invention may include a step c) of subculturing and culturing the cell population cultured in step b) in a predetermined medium.
- the medium in step c) of the method for producing a cell population of the present invention contains IL-2.
- the medium in step c) may further contain a 4-1BB agonist, or may further contain IL-21.
- IL-2, 4-1BB agonist, and IL-21 please refer to the above.
- the medium in step c) of the present invention contains IL-2, but does not contain IL-21 or a 4-1BB agonist.
- the medium in step c) of the present invention contains IL-2, IL-21, and a 4-1BB agonist.
- the concentration of IL-21 in the medium can be, for example, 0.01 ng/mL or more, 0.1 ng/mL or more, or 1 ng/mL or more, or 100 ng/mL or less, 10 ng/mL or less, or 5 ng/mL or less.
- the concentration of the 4-1BB agonist in the medium can be, for example, 0.01 ⁇ g/mL or more, 0.1 ⁇ g/mL or more, or 1 ⁇ g/mL or more, or 100 ⁇ g/mL or less, 10 ⁇ g/mL or less, or 5 ⁇ g/mL or less.
- the concentration of the anti-4-1BB antibody in the medium in step c) can be 0.01 ⁇ g/mL or more, preferably 0.05 ⁇ g/mL or more, more preferably 0.1 ⁇ g/mL or more, even more preferably 0.5 ⁇ g/mL or more, and particularly preferably 1 ⁇ g/mL or more, or may be 100 ⁇ g/mL or less, 10 ⁇ g/mL or less, 5 ⁇ g/mL or less, 4 ⁇ g/mL or less, 3 ⁇ g/mL or less, or 2 ⁇ g/mL or less.
- the concentration of 4-1BBL in the medium in step c) can be 0.1 ⁇ g/mL or more, preferably 0.5 ⁇ g/mL or more, more preferably 1 ⁇ g/mL or more, even more preferably 5 ⁇ g/mL or more, and particularly preferably 10 ⁇ g/mL or more.
- Step c) may include culturing the cell population cultured in step b) for a predetermined period of time.
- the predetermined period is 1 day or more, and may be 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, or 30 days or less, 25 days or less, 20 days or less, 15 days or less, 14 days or less, 13 days or less, 12 days or less, 11 days or less, or less, and is not particularly limited.
- the culture period in step c) is preferably 4 to 10 days, more preferably 5 to 9 days, even more preferably 6 to 8 days, and particularly preferably 7 days.
- the cells may be added to the same medium, or the medium may be replaced (in this case, it can be said that step c) is continued as long as the medium meets the requirements).
- the method for producing a cell population according to the present invention may include a step of further culturing the cell population cultured in step c) under the same conditions as in step c), i.e., repeating step c).
- Step c) may be repeated multiple times, with no limit on the number of times, as long as cell proliferation is achieved.
- the number of cells contained in the cell population obtained by removing CD3-positive cells and CD34-positive cells from the cell population containing mononuclear cells used in step a) is amplified at least 2-fold, preferably 10-fold, and more preferably 100-fold.
- the cell population obtained by the production method of the present invention is at least CD3 negative and CD56 positive, and preferably contains cells having a phenotype of one or more cell surface markers selected from the group consisting of CD3 negative, CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 (Integrin ⁇ 1) positive, and ITGA3 (Integrin ⁇ 3) positive. More preferably, the cell population obtained by the production method of the present invention contains cells that are CD3 negative, CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, and ITGA3 positive.
- CCR or CXCR used as a cell surface marker is a type of chemokine receptor, and ITG (Integrin) is a type of cell adhesion factor.
- the proportion of CD3-negative and CD56-positive cells in the cell population may be, for example, 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, preferably 60% or more, more preferably 70% or more, even more preferably 80% or more, and particularly preferably 90% or more.
- a cell population in which the proportion of CD3-negative and CD56-positive cells in the cell population is lower than any of the above proportions is distinguished from a cell population containing CD3-negative and CD56-positive cells obtained by the present invention.
- the percentage of cells having each phenotype of cell surface marker in the cell population may be, for example, 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, preferably 60% or more, more preferably 70% or more, even more preferably 80% or more, and particularly preferably 90% or more.
- a cell population in which the proportion of cells that are CD3 negative and CD56 positive, and CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, or ITGA3 positive is less than any of the above proportions, is distinguished from a cell population obtained by the present invention that contains cells that are CD3 negative and CD56 positive, and CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, or ITGA3 positive.
- the CD3 negative and CD56 positive cells in the cell population obtained by the present invention are preferably CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, and ITGA3 positive, and more preferably CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, and ITGA3 positive.
- the cells contained in the cell population obtained by the present invention can exhibit high cytotoxic activity.
- the cytotoxic activity can be evaluated, for example, by measuring the lysis ability of target cells (effector cells (E)) against target cells (T) such as tumor cells.
- target cells effector cells (E)
- T target cells
- tumor cell lines of leukemia, glioma, breast cancer, colon cancer, ovarian cancer, prostate cancer, etc. can be used as tumor cells, but are not limited thereto.
- spheroids prepared from tumor cell lines can also be used as target cells.
- the ratio of effector cells to target cells (E:T) is not particularly limited, and may be, for example, 1:1, 2:1, 5:1, etc.
- the cytotoxic activity can be expressed as the percentage (%) of target cells killed by effector cells, and is calculated by the following formula.
- Cytotoxic activity can be measured, for example, by distinguishing between effector cells and target cells, and between live and dead cells, using reagents such as antibodies labeled with radioactive substances or fluorescent dyes, quantifying these cells using flow cytometry or the like, and determining the cell death rate according to the above formula.
- the incubation time with target cells can be set appropriately depending on the type of target cells used; for example, when K562 cells, a leukemia cell line, are used as target cells, it can be set to 2 hours.
- high cytotoxic activity means that the cytotoxic activity is 50% or more when the target cells are K562 cells, mixed at an E:T ratio of 1-5:1, and co-cultured for 2 hours.
- the cytotoxic activity is preferably 60% or more, and more preferably 70% or more.
- the desired cytotoxic activity can also be determined by detecting CD107a-positive cells in the effector cells. Since CD107a is present in granules in NK cells and migrates to the cell membrane surface of NK cells upon degranulation, CD107a positivity indirectly indicates that cytotoxic activity, i.e., attack of target cells by cells contained in the cell population obtained by the present invention, has been obtained. CD107a-positive cells can be detected, for example, by measuring luminescence after incubating target cells with a fluorescently labeled anti-CD107a antibody.
- the proportion of CD107a-positive cells in the cell population obtained by the present invention is at least 20% or more, preferably 30% or more, more preferably 40% or more, and even more preferably 50% or more.
- the tumor cell line may be, for example, a leukemia cell line, such as K562 cells.
- the cell population obtained by the present invention exerts an antitumor effect even when administered in vivo.
- the antitumor effect in vivo can be evaluated, for example, by administering the cell population obtained by the present invention to a cancer-bearing model animal and measuring the change in tumor volume or the number of tumor cells.
- the preparation of the cancer-bearing model animal and the measurement of the change in tumor volume or the number of tumor cells can be performed using methods known to those skilled in the art, for example, a method of transplanting fluorescently labeled tumor cells into the animal and observing them.
- a specific example is a method using an in vivo imaging system (IVIS) in which luciferase-expressing SKOV cells (human ovarian cancer cell line) are transplanted into an immunodeficient mouse, luciferin is administered to the mouse, and the tumor luminescence is observed under anesthesia.
- IVIS in vivo imaging system
- the administration method of the cell population obtained by the present invention is not particularly limited, and the number of administrations may be single or multiple, and the administration interval when multiple administrations are performed can be appropriately set.
- the timing of administration can be any time point, for example, 1 to 30 days after tumor transplantation.
- the dosage may be, for example, 1.0 ⁇ 10 4 to 1.0 ⁇ 10 10 cells/body per administration, preferably 1.0 ⁇ 10 5 to 1.0 ⁇ 10 9 cells/body, more preferably 1.0 ⁇ 10 6 to 1.0 ⁇ 10 8 cells/body, and particularly preferably 1.5 ⁇ 10 7 to 2.7 ⁇ 10 7 cells/body.
- the route of administration may be any, including intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration, with intraperitoneal administration being preferred.
- IL-2 may be administered simultaneously in order to maintain the activity of the cell population obtained by the present invention in vivo.
- the dosage of IL-2 in this case may be 1,000 to 10,000 IU/body, and preferably 5,000 IU/body.
- administration of the cell population obtained by the present invention can reduce the number of tumor cells in a cancer-bearing model animal to 1/10 or less, compared to the case where the cell population is not administered.
- PBMC Cryopreserved PBMCs Peripheral Blood Mononuclear Cells
- RPMI 1640 medium containing 10% FBS and 1% penicillin-streptomycin mixed solution
- PBMC (CD3 ⁇ , CD34 ⁇ ) 4x10 5 cells/mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1 ng/mL]
- PBMC (CD3 ⁇ , CD34 ⁇ ) 4x10 5 cells/mL, RPMI medium *2 , Urelumab [1 ⁇ g/mL], IL-21 [1 ng/mL]
- PBMC (CD3 + , CD34 + ) 4x10 5 cells/mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1 ng/mL]
- PBMC (CD3 + , CD34 + ) 4x10 5 cells/mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1 ng/mL]
- PBMC (CD3 + , CD34 + ) 4x10 5 cells/m
- (3)-5-1 and (3)-5-2 were each seeded in 3.0 mL portions into a 6-well plate under the following conditions, and subcultured.
- (3)-5-1 (3)-5-1 Cell suspension 5x10 5 cells/3 mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1 ng/mL] (3)-5-2-a (3)-5-2 cell suspension 5x10 5 cells/3mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1ng/mL] (3)-5-2-b (3)-5-2 cell suspension 1.5x10 6 cells/3 mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1 ng/mL]
- Figure 2 shows the change in cell proliferation rate. It was found that the cell proliferation efficiency was better when NTI medium was used than when RPMI medium was used.
- the results of flow cytometry performed on each cell surface marker for (1), (3), and (4) are shown in Figure 3.
- the obtained cell population contained many cells with a phenotype similar to the cells exhibiting high cytotoxicity described in Patent Document 1, etc., but CD3-positive cells were also amplified.
- Example 3 (Donor Mix B) A culture test was carried out using different concentrations of anti-4-1BB antibody and IL-21.
- PBMC CD3- , CD34- 4x105 cells/mL, KBM502 medium *3
- PBMC CD3 ⁇ , CD34 ⁇ 4x10 5 cells/mL, KBM502 medium *3 , IL-21 [1 ng/mL]
- PBMC CD3 ⁇ , CD34 ⁇ 4x10 5 cells/mL, KBM502 medium *3 , IL-21 [10 ng/mL]
- PBMC CD3 ⁇ , CD34 ⁇ 4x10 5 cells/mL, KBM502 medium *3 , IL-21 [10 ng/mL]
- PBMC CD3 ⁇ , CD34 ⁇ 4x10 5 cells/mL, KBM502 medium *3 , IL-21 [100 ng/mL]
- PBMC CD3 ⁇ , CD34 ⁇ 4x10 5 cells/mL, KBM502 medium *3 , IL-21 [100 ng/mL]
- PBMC CD3
- ⁇ K562 ⁇ K562 cells were suspended in RPMI1640 medium (serum-free) and stained using PKH26 Red Fluorescent Cell Linker Kit, and then adjusted to 2 x 10 6 cells/mL in RPMI1640 medium (containing 10% FBS and 1% penicillin-streptomycin mixed solution).
- Group composition As shown in the table below, the test was conducted on two groups: a control (untreated) group and a test substance administration group.
- Example 6 (Donor Mix C) ⁇ Day 0 Cryopreserved PBMC (CD3 ⁇ , CD34 ⁇ ) were thawed by a predetermined method, and culture was initiated under the following conditions. (1) 1.3x105 cells/0.65mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1ng/mL], 24-well plate (2) 1.6x105 cells/0.65mL, KBM501 medium *4 , 24-well plate (3) 1.3x105 cells/0.65mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1ng/mL], G-Rex 24-well plate (4) 1.6x105 cells/0.65mL, KBM501 medium *4 , G-Rex 24-well plate A 7.5x106 cells/15mL, KBM501 medium *4 , T75 flask B 7.5x10 6 cells/15mL, KBM501 medium *4 , T75 flask
- the G-Rex 24-well plate was thoroughly pipetted, the number of cells was counted, and 4 mL of fresh NTI medium *1 was added and the cells were subcultured.
- T75 flasks A and B were thoroughly pipetted, and the cells were harvested using 1 mM EDTA/PBS and counted.
- the number of viable cells was counted, and 2x108 cells were suspended in 5mL of HSC-BANKER (ZENOAQ, CB071) and frozen at -80°C.
- the frozen cells were thawed in a water bath at 37°C, and then diluted 10-fold with Plasma-Lyte A.
- the cytotoxic activity rate is evaluated as A to D as follows. When the evaluation is C or higher, it is determined that high cytotoxic activity is obtained and the purpose is achieved.
- Example 7 (Donor Mix C, Single Donor A) ⁇ Day 0 Cryopreserved PBMCs were thawed by a prescribed method, and 1.8 mL of the cell suspension prepared for the following five groups was seeded onto a 6-well plate to initiate culture.
- PBMC (CD3 ⁇ , CD34 ⁇ ) 4x10 5 cells/mL, KBM501 medium *4 , Urelumab [1 ⁇ g/mL], IL-21 [1 ng/mL]
- PBMC (CD3 ⁇ , CD34 ⁇ ) 4x10 5 cells/mL, NTI medium *1 , Urelumab [1 ⁇ g/mL], IL-21 [1 ng/mL]
- PBMC ( CD3- , CD34- ) 4x105 cells/mL, Simulect, KBM501 medium containing Prograf *5 , Urelumab [1 ⁇ g/mL], IL-21 [1ng/mL]
- PBMC ( CD3- , CD34- ) 4x105 cells/mL, Simulect, NTI medium containing Prograf *6 , Urelumab [1 ⁇ g/mL], IL-21 [1ng/mL]
- PBMC (CD3 + , CD
- (2)-1 Dilute to 5x105 cells/3mL with new NTI medium *4 , add Urelumab [1 ⁇ g/mL] and IL-21 [1ng/mL] and sow.
- (2)-2 Dilute to 5x105 cells/3mL with new NTI medium *1 and sow.
- (2)-3 Dilute to 5x105 cells/1.83mL with new NTI medium *1 and sow.
- (2)-4 Take 5x105 cells, centrifuge at 500g for 5 minutes, then suspend well in 3mL of new NTI medium *1 containing Urelumab [1 ⁇ g/mL] and IL-21 [1ng/mL] and sow.
- (5)-1 Dilute to 5x105 cells/3mL with new NTI medium *1 , add Urelumab [1 ⁇ g/mL] and IL-21 [1ng/mL] and sow.
- (5)-2 Dilute to 5x105 cells/3mL with new NTI medium *1 and sow.
- (5)-3 Dilute to 5x105 cells/1.83mL with new NTI medium *1 and sow.
- (5)-4 Take 5x105 cells, centrifuge at 500g for 5 minutes, then suspend well in 3mL of new NTI medium *1 containing Urelumab [1 ⁇ g/mL] and IL-21 [1ng/mL] and sow.
- Each of the wells (1)-3, (2)-3, (5)-3, and (4)-5 was subcultured in 2 mL of 5-fold dilution (0.4 mL of cell suspension and 1.6 mL of new medium) and 10-fold dilution (0.2 mL of cell suspension and 1.8 mL of new medium) onto a 6-well plate.
- Example 8 Cryopreserved PBMCs were completely thawed in a 37°C water bath and diluted 10-fold with KBM501/5% UG medium *7 . After centrifugation at 500 ⁇ g for 5 minutes, the cells were adjusted to 4 ⁇ 10 5 cells/mL with the following 6 groups of medium, and 0.8 mL was seeded on a 12-well plate to start culture.
- the cells were centrifuged (500 x g, 5 minutes), the supernatant was removed, and the cells were suspended in PBS. Measurements were then performed using a flow cytometer (BD LSRFortessa, BD Biosciences) and analyzed using FlowJo software (FLOWJO, LLC).
- NTI (FUKOKU, T2108251) containing 5% UltraGRO (AventaCell, HPCPLCRL10) and 2U/mL heparin sodium (Nipro), Simulect 0.5 ⁇ g/mL, and Prograf 1ng/mL
- Alexa Fluor® 700-labeled anti-human CD56 antibody (Biolegend, 318316) ⁇ APC-labeled anti-human NKp30 antibody (Biolegend, 325210) PerCP/Cy5.5-labeled anti-human CD3 antibody (Biolegend, 300430) PE-Cy7-labeled anti-human CD16 antibody (Biolegend, 302016) PE-labeled anti-human CD19 antibody (Biolegend, 302208) FITC-labeled anti-human CD14 antibody (Biolegend, 325604) APC-Cy7-labeled anti-human CD34 antibody (Biolegend, 343514)
- FITC-labeled anti-human CD49a antibody Biolegend, 328307) PE-labeled anti-human CD49c antibody (Biolegend, 343803) APC-labeled anti-human CD61 antibody (Biolegend, 336412) PE-Dazzle594-labeled anti-human NKp30 antibody (Biolegend, 325231) PE-Cy7-labeled anti-human CCR5 antibody (Biolegend, 359107) Pacific BlueTM-labeled anti-human CCR6 antibody (Biolegend, 353438) APC-Cy7-labeled anti-human CXCR3 antibody (Biolegend, 353721)
Landscapes
- Engineering & Computer Science (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Wood Science & Technology (AREA)
- Biotechnology (AREA)
- Organic Chemistry (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biomedical Technology (AREA)
- Genetics & Genomics (AREA)
- General Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- General Health & Medical Sciences (AREA)
- Virology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Medicinal Chemistry (AREA)
- Cell Biology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
本発明は、CD3陰性かつCD56陽性である細胞を含む細胞集団の製造方法または増幅活性化方法に関する。 The present invention relates to a method for producing or amplifying and activating a cell population containing CD3-negative and CD56-positive cells.
ナチュラルキラー細胞(NK細胞)は、CD3陰性、CD56陽性等の表面マーカーの表現型を有し、自然免疫の主要因子として働く細胞傷害性リンパ球の一種である。 Natural killer cells (NK cells) are a type of cytotoxic lymphocyte that have a surface marker phenotype of CD3 negative and CD56 positive, and act as a key factor in innate immunity.
キメラ抗原受容体(CAR)発現T細胞(CAR−T細胞)を用いたCAR−T療法が、再発または難治性のCD19陽性B細胞性急性リンパ芽球性白血病等を適応症として承認されているが、CAR−T療法に代わる新世代治療技術として、悪性腫瘍の治療製剤への高活性NK細胞の応用の可能性が考えられている(非特許文献1)。これまでに、固形腫瘍に高い傷害活性を示す細胞の集団の製造方法が知られている(特許文献1、非特許文献2)。 CAR-T therapy using chimeric antigen receptor (CAR)-expressing T cells (CAR-T cells) has been approved for the treatment of relapsed or refractory CD19-positive B-cell acute lymphoblastic leukemia, etc., but the application of highly active NK cells to therapeutic preparations for malignant tumors is considered to be a new generation therapeutic technique to replace CAR-T therapy (Non-Patent Document 1). Methods for producing cell populations that exhibit high cytotoxicity against solid tumors are known (Patent Document 1, Non-Patent Document 2).
また、胚性幹細胞(ES細胞)/人工多能性幹細胞(iPS細胞)由来のNK細胞製剤候補も報告されている。例えば、非特許文献3には、NK2GDの膜貫通ドメイン、2B4(CD244)共刺激ドメインおよびCD3ζシグナルドメインを含むCARを発現する、ヒトiPS細胞由来のNK細胞が記載されている。 Also, candidate NK cell preparations derived from embryonic stem cells (ES cells)/induced pluripotent stem cells (iPS cells) have been reported. For example, Non-Patent Document 3 describes NK cells derived from human iPS cells that express a CAR containing the transmembrane domain of NK2GD, the 2B4 (CD244) costimulatory domain, and the CD3ζ signal domain.
他方で、末梢血単核細胞からリンパ球を増幅させる手段として、4−1BBリガンド(4−1BBL)やOX40リガンド、インターロイキン(IL)−18、IL−21等を用いる方法が知られる(非特許文献4)。特許文献2には、4−1BBアゴニスト等の腫瘍壊死因子受容体スーパーファミリーアゴニストを使用して腫瘍浸潤リンパ球を拡大培養する方法が記載されている。特許文献3には、IL−2、IL−15およびIL−21をそれぞれ所定の濃度で含み、液状である、T細胞を増殖させるための組成物が記載されている。 On the other hand, methods using 4-1BB ligand (4-1BBL), OX40 ligand, interleukin (IL)-18, IL-21, etc. are known as means for amplifying lymphocytes from peripheral blood mononuclear cells (Non-Patent Document 4). Patent Document 2 describes a method for expanding tumor-infiltrating lymphocytes using tumor necrosis factor receptor superfamily agonists such as 4-1BB agonists. Patent Document 3 describes a liquid composition for expanding T cells, which contains IL-2, IL-15, and IL-21 at predetermined concentrations.
一方で、非特許文献4には、実験した条件では、4−1BBLまたはIL−21を液性因子として用いる場合には、これらをビーズに固定化して用いる場合と比較して、NK細胞の増幅効果が劣ることが記載されていることから、4−1BBL、IL−21等の因子を液性因子として用いるのは非効率的であることが示唆される。そこで、遺伝子組換えfeeder細胞株や合成脂質膜を用いることで、NK細胞の高い増殖効率を実現する手段が開発されている。例えば、非特許文献5には、膜結合型IL−21を発現する遺伝子組換え人工抗原提示細胞とNK細胞を共培養する方法が記載されている。特許文献4には、CD3陽性細胞が除去された末梢血液単核細胞をfeeder細胞と共に増殖させ、末梢血単核細胞が特定の累積分裂レベルに到達する時点で、feeder細胞で再刺激する、NK細胞の生産方法が記載されている。 On the other hand, Non-Patent Document 4 describes that under the experimental conditions, when 4-1BBL or IL-21 is used as a humoral factor, the NK cell expansion effect is inferior compared to when these are immobilized on beads, suggesting that using factors such as 4-1BBL and IL-21 as humoral factors is inefficient. Therefore, a means for realizing high NK cell proliferation efficiency by using a genetically modified feeder cell line or a synthetic lipid membrane has been developed. For example, Non-Patent Document 5 describes a method for co-culturing NK cells with genetically modified artificial antigen presenting cells expressing membrane-bound IL-21. Patent Document 4 describes a method for producing NK cells, in which peripheral blood mononuclear cells from which CD3 positive cells have been removed are grown together with feeder cells, and when the peripheral blood mononuclear cells reach a specific cumulative division level, the cells are restimulated with feeder cells.
従来の方法によって得られる細胞は、固形腫瘍に高い傷害活性を示す一方で、その供給量は限定的である。 While cells obtained by conventional methods show high cytotoxicity against solid tumors, their supply is limited.
非特許文献3のようなES細胞/iPS細胞由来のNK細胞を用いた製剤においては、特に固形腫瘍に対する活性が十分ではない。また、特許文献2または特許文献3に記載される発明は、T細胞の増殖に関するものであり、NK細胞の増殖に有用であることは示されていない。非特許文献5や特許文献4のような遺伝子組換え技術やfeeder細胞株を用いる方法においては、品質管理のハードルが上がるうえに、製造/精製工程が煩雑になり(非特許文献6)、さらに医療費負担の拡大も課題となり得る。 In preparations using NK cells derived from ES cells/iPS cells such as those in Non-Patent Document 3, activity against solid tumors is particularly insufficient. Furthermore, the inventions described in Patent Documents 2 and 3 relate to the proliferation of T cells, and have not been shown to be useful for the proliferation of NK cells. In methods using gene recombination techniques or feeder cell lines such as those in Non-Patent Document 5 and Patent Document 4, the hurdles of quality control are increased, the manufacturing/purification processes become complicated (Non-Patent Document 6), and the burden of medical expenses may also increase.
そこで、本発明は、feeder細胞株を用いず、かつ、遺伝子組換え技術を使用せずに、CD3陰性かつCD56陽性である細胞を含む細胞集団を効率的に増殖させることを目的とする。 The present invention aims to efficiently grow a cell population containing CD3-negative and CD56-positive cells without using a feeder cell line or genetic recombination techniques.
本発明者らは上記課題を解決するために鋭意検討したところ、a)単核球を含む細胞集団からCD3陽性細胞およびCD34陽性細胞を除去した細胞集団を、IL−2、IL−21および4−1BBアゴニストを含む培地中で培養する工程であって、IL−21および4−1BBアゴニストが液性因子である、工程を含む方法により、CD3陰性かつCD56陽性である細胞を含む細胞集団を、従来の製造方法と比較して大量に増幅して得ることができることを見出し、本発明を完成させるに至った。 The inventors conducted extensive research to solve the above problems and discovered that a method including the steps of: a) culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a medium containing IL-2, IL-21 and a 4-1BB agonist, where the IL-21 and the 4-1BB agonist are humoral factors, can mass-produce and amplify a cell population containing CD3-negative and CD56-positive cells, as compared to conventional manufacturing methods, and thus completed the present invention.
すなわち、本発明は以下に関する。
[1]以下の工程を含む、CD3陰性かつCD56陽性である細胞を含む細胞集団の製造方法。
a)単核球を含む細胞集団からCD3陽性細胞およびCD34陽性細胞を除去した細胞集団を、IL−2、IL−21および4−1BBアゴニストを含む培地中で1日以上培養する工程であって、IL−21および4−1BBアゴニストが液性因子である、工程。
[2]以下の工程をさらに含む、[1]に記載の製造方法。
b)工程a)で培養した細胞集団を、IL−2および4−1BBアゴニストを含み、IL−21を含まない培地で継代し、1日以上培養する工程。
[3]工程a)の培地中の4−1BBアゴニストの濃度が1μg/mL以上であり、工程a)の培地中のIL−21の濃度が1ng/mL以上である、[1]または[2]に記載の製造方法。
[4]以下の工程をさらに含む、[2]に記載の製造方法。
c)工程b)で培養した細胞集団を、IL−2を含む培地で継代し、1日以上培養する工程。
[5]4−1BBアゴニストが、抗4−1BB抗体および4−1BBリガンドから選択される、[1]~[4]のいずれか1項に記載の方法。
[6]培地が血小板溶解物を含む、[1]~[5]のいずれか1項に記載の方法。
That is, the present invention relates to the following.
[1] A method for producing a cell population containing CD3-negative and CD56-positive cells, comprising the steps of:
a) culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a medium containing IL-2, IL-21 and a 4-1BB agonist for one day or more, wherein the IL-21 and the 4-1BB agonist are humoral factors.
[2] The method according to [1], further comprising the steps of:
b) Passaging the cell population cultured in step a) in a medium containing IL-2 and a 4-1BB agonist but not containing IL-21, and culturing the medium for one day or more.
[3] The method for production according to [1] or [2], wherein the concentration of the 4-1BB agonist in the medium in step a) is 1 μg/mL or more, and the concentration of IL-21 in the medium in step a) is 1 ng/mL or more.
[4] The method according to [2], further comprising the steps of:
c) Passaging the cell population cultured in step b) in a medium containing IL-2 and culturing for one day or more.
[5] The method according to any one of [1] to [4], wherein the 4-1BB agonist is selected from an anti-4-1BB antibody and a 4-1BB ligand.
[6] The method according to any one of [1] to [5], wherein the medium contains a platelet lysate.
本発明によれば、feeder細胞株を用いず、かつ、遺伝子組換え技術を使用しなくても、CD3陰性かつCD56陽性である細胞を含む細胞集団を、従来の製造方法と比較して、大量に増幅して得ることができる。 According to the present invention, a cell population containing CD3-negative and CD56-positive cells can be mass-multiplied and obtained without using a feeder cell line or genetic recombination techniques, as compared to conventional manufacturing methods.
[細胞集団の製造方法]
<工程a>
本発明の一態様においては、以下の工程を含む、CD3陰性かつCD56陽性である細胞を含む細胞集団の製造方法が提供される。
a)単核球を含む細胞集団からCD3陽性細胞およびCD34陽性細胞を除去した細胞集団を、所定の培地中で培養する工程。
[Method of producing cell population]
<Step a>
In one aspect of the present invention, there is provided a method for producing a cell population containing CD3-negative and CD56-positive cells, the method comprising the steps of:
a) a step of culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a predetermined medium.
<細胞集団>
本発明において、細胞集団とは、複数個の細胞、例えば1×105cells以上の細胞で構成される一群をいう。本発明により提供される細胞集団は、様々な細胞密度に調製することができる。例えば、1×105cells/mL以上に調製することができる。
<Cell population>
In the present invention, a cell population refers to a group of cells consisting of a plurality of cells, for example, 1×10 5 cells or more. The cell population provided by the present invention can be prepared to various cell densities. For example, it can be prepared to 1×10 5 cells/mL or more.
本発明においては、少なくともCD3陰性かつCD56陽性である細胞を含む細胞集団が得られる。CD3陰性かつCD56陽性であるか否かは、細胞表面マーカーの発現パターンを分析することにより判断することができる。本発明において、細胞表面マーカーが陽性であるか否かは、例えば、フローサイトメトリー法により、対象の細胞表面マーカーを発現しないかまたは低発現である対照の細胞集団を用いて、細胞表面マーカーによるシグナルを比較することにより判断することができる。細胞表面マーカーが陰性であるか否かについても、同様の手法により判断することができる。 In the present invention, a cell population containing at least CD3-negative and CD56-positive cells is obtained. Whether or not a cell is CD3-negative and CD56-positive can be determined by analyzing the expression pattern of the cell surface marker. In the present invention, whether or not a cell surface marker is positive can be determined, for example, by flow cytometry, by comparing the signal from the cell surface marker with that of a control cell population that does not express or expresses the target cell surface marker at a low level. Whether or not a cell surface marker is negative can also be determined by a similar method.
本発明の細胞集団の製造方法には、単核球を含む細胞集団からCD3陽性細胞およびCD34陽性細胞を除去した細胞集団を用いることができる。単核球を含む細胞集団は、T細胞、B細胞、NK細胞、単球、樹状細胞等の多様なリンパ球を含み得る。単核球を含む細胞集団は、ドナーから採取した血球細胞から単核球を分離することにより得られる末梢血単核細胞であってよい。血球細胞は、当業者に公知の方法によってドナーから採取することができ、例えば、アフェレーシス法により、ドナーの末梢血から採取することができる。血球細胞は、ドナーの臍帯血、骨髄、リンパ節等から採取したものであってもよい。血球細胞からの単核球の分離は、例えば、密度勾配遠心法を用いて行うことができる。血球細胞は、胚性幹細胞(ES細胞)や人工多能性幹細胞(iPS細胞)を分化させて得たものを用いてもよい。血球細胞に分化させる方法は、当業者に公知の方法によって行うことができ、例えば、H.Melichar et al.Comparative Study of Hematopoietic Differentiation between Human Embryonic Stem Cell Lines.PLoS One.2011;6(5):e19854.doi:10.1371/journal.pone.0019854.等に記載された手法を参照することができる。単核球を含む細胞集団からのCD3陽性細胞およびCD34陽性細胞の除去は、当業者に公知の方法に従って行ってよく、例えば、CD3またはCD34に対する抗体が結合した磁気ビーズを用いて、CD3陽性細胞またはCD34陽性細胞を分離する方法により行うことができる。 In the method for producing a cell population of the present invention, a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing monocytes can be used. The cell population containing monocytes can contain various lymphocytes such as T cells, B cells, NK cells, monocytes, and dendritic cells. The cell population containing monocytes can be peripheral blood mononuclear cells obtained by separating monocytes from blood cells collected from a donor. Blood cells can be collected from a donor by a method known to those skilled in the art, for example, by apheresis from the donor's peripheral blood. Blood cells may be collected from the donor's umbilical cord blood, bone marrow, lymph nodes, etc. The separation of mononuclear cells from blood cells can be performed, for example, by density gradient centrifugation. Blood cells obtained by differentiating embryonic stem cells (ES cells) or induced pluripotent stem cells (iPS cells) can also be used. The method for differentiating into blood cells can be performed by a method known to those skilled in the art, for example, by apheresis from the donor's peripheral blood. The methods described in Melichar et al. Comparative Study of Hematopoietic Differentiation between Human Embryonic Stem Cell Lines. PLoS One. 2011; 6(5): e19854. doi: 10.1371/journal. pone. 0019854. and the like can be referred to. Removal of CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells may be performed according to a method known to those skilled in the art, for example, by a method of separating CD3-positive cells or CD34-positive cells using magnetic beads bound to an antibody against CD3 or CD34.
単核球を含む細胞集団は、単独のドナーに由来するものであっても、複数のドナーに由来するものであってもよい。単核球を含む細胞集団が複数のドナーに由来する場合、細胞集団中、各ドナー由来の細胞集団の割合は、特に限定されない。 The cell population containing mononuclear cells may be derived from a single donor or from multiple donors. When the cell population containing mononuclear cells is derived from multiple donors, the proportion of the cell population derived from each donor in the cell population is not particularly limited.
単核球を含む細胞集団は、凍結保存していたものを解凍して使用してもよい。凍結保存は、例えば、ジメチルスルホキシド(DMSO)等の凍結保護試薬を含んだ完全培地等の凍結保存に適した培地を用いて、適切な細胞密度になるように単核球を含む細胞集団を懸濁し、クライオバイアル等の凍結保存用の容器に入れて、−80℃以下にて保管することにより行うことができる。解凍は、例えば、単核球を含む細胞集団の入った凍結保存用容器を37℃の水浴に移し、緩やかに旋回させることにより行うことができる。 The cell population containing mononuclear cells may be used after being thawed from cryopreservation. Cryopreservation can be performed, for example, by suspending the cell population containing mononuclear cells to an appropriate cell density in a medium suitable for cryopreservation, such as a complete medium containing a cryoprotective agent such as dimethyl sulfoxide (DMSO), placing it in a cryopreservation container such as a cryovial, and storing it at -80°C or below. Thawing can be performed, for example, by transferring the cryopreservation container containing the cell population containing mononuclear cells to a 37°C water bath and gently swirling it.
<培地>
本発明の細胞集団の製造方法は、単核球を含む細胞集団からCD3陽性細胞およびCD34陽性細胞を除去した細胞集団を、所定の培地中で培養する工程a)を含む。培地には、培養に適する添加物、試薬等がさらに含まれていてもよい。以下、「本発明の細胞集団の製造方法における培地」、「本発明の培地」、「培地」等という場合であって、特に「工程a)における培地」または「工程b)における培地」等と特定しない場合には、工程a)、工程b)および工程c)から選択される1以上における培地を指すものとする。
<Culture medium>
The method for producing a cell population of the present invention includes a step a) of culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a predetermined medium. The medium may further contain additives, reagents, etc. suitable for culture. Hereinafter, when referring to "medium in the method for producing a cell population of the present invention", "medium of the present invention", "medium" or the like, and not particularly specifying "medium in step a)" or "medium in step b"), etc., it refers to the medium in one or more selected from step a), step b) and step c).
本明細書において、以下にサイトカイン、ポリペプチド、添加物等の培地中の濃度を記載する場合、その濃度は、工程a)、工程b)、工程c)等の各工程の開始時における培地中の濃度であればよく、各工程の期間全体にわたる濃度を規定するものではない。 In this specification, when the concentration of cytokines, polypeptides, additives, etc. in the medium is described below, the concentration refers to the concentration in the medium at the start of each step, such as step a), step b), step c), etc., and does not specify the concentration throughout the entire period of each step.
(IL−2、IL−21、4−1BBアゴニスト)
本発明の細胞集団の製造方法における培地は、前記細胞集団を増幅するのに適切なサイトカイン、ポリペプチド等を含んでいてもよい。適切なサイトカインは、例えば1種以上のインターロイキン(IL)であり、好ましくはIL−2およびIL−21である。適切なポリペプチドは、好ましくは4−1BBアゴニストである。4−1BBは、腫瘍壊死因子受容体スーパーファミリー(TNFRSF)に属し、主にリンパ球の細胞表面に発現する膜貫通型受容体である。4−1BBは、CD137、TNFRSF9等と称されることもある。本発明において、例えば4−1BBアゴニストという場合、4−1BBに結合してその機能を促進することができるものをいう。4−1BBの機能としては、例えば、ヒト末梢血単核細胞からのIL−8の産生やヒトTリンパ球からのインターフェロン(IFN)−γの産生が挙げられ、4−1BBによるIL−8やIFN−γの産生は、当業者に公知の方法によって測定することが可能である。4−1BBアゴニストは、例えば、4−1BBに結合し、IL−8やIFN−γの産生を促進することができるものであってよい。4−1BBアゴニストとしては、例えば、抗4−1BB抗体、4−1BBリガンド(4−1BBL)等が挙げられる。
(IL-2, IL-21, 4-1BB agonists)
The medium in the method for producing a cell population of the present invention may contain a cytokine, polypeptide, etc. suitable for expanding the cell population. Suitable cytokines are, for example, one or more interleukins (IL), preferably IL-2 and IL-21. Suitable polypeptides are preferably 4-1BB agonists. 4-1BB belongs to the tumor necrosis factor receptor superfamily (TNFRSF) and is a transmembrane receptor that is mainly expressed on the cell surface of lymphocytes. 4-1BB is also called CD137, TNFRSF9, etc. In the present invention, for example, a 4-1BB agonist refers to one that can bind to 4-1BB and promote its function. Examples of the function of 4-1BB include the production of IL-8 from human peripheral blood mononuclear cells and the production of interferon (IFN)-γ from human T lymphocytes, and the production of IL-8 and IFN-γ by 4-1BB can be measured by a method known to those skilled in the art. The 4-1BB agonist may be, for example, one that can bind to 4-1BB and promote the production of IL-8 or IFN-γ. Examples of the 4-1BB agonist include anti-4-1BB antibodies and 4-1BB ligand (4-1BBL).
本明細書において、抗4−1BB抗体という場合、4−1BBにおける1以上の特定の抗原決定基を認識し、これに結合し、4−1BBを多量体化することができる抗体をいい、例えば、ウレルマブ(Urelumab)、ウトミルマブ(Utomilumab)、またはこれらと同じ抗原決定基を認識することができる抗体が挙げられる。 In this specification, anti-4-1BB antibodies refer to antibodies that can recognize and bind to one or more specific antigenic determinants in 4-1BB and can multimerize 4-1BB, such as urelumab, utomilumab, or antibodies that can recognize the same antigenic determinants as these.
本明細書において、4−1BBリガンド(4−1BBL)という場合、4−1BBに結合することができるポリペプチドやタンパク質をいう(但し、抗体を除く)。4−1BBLは、当業者に公知の方法によって合成されたものであってよく、遺伝子組み換え技術によって人工的に作製されたものであってもよく、遺伝子組み換え技術によって人工的に作製された4−1BBLをリコンビナント4−1BBLという。一つの態様において、4−1BBLは、以下に示す配列番号:1もしくは配列番号:2のアミノ酸配列からなるポリペプチド、4−1BBLの細胞外ドメイン(配列番号:3)を含むポリペプチド、または配列番号:1、配列番号:2もしくは配列番号:3と少なくとも70%以上、80%以上、85%以上、90%以上、95%以上、96%以上、97%以上、98%以上もしくは99%以上の同一性を有するアミノ酸配列からなり、かつCD3陰性かつCD56陽性である細胞の増殖活性を有するポリペプチドであってよい。アミノ酸配列の同一性とは、対象のポリペプチド同士のアミノ酸配列の同一性をいい、アミノ酸配列の最適なアラインメントにおいて一致するアミノ酸残基の割合(%)によって表されるものをいう。アミノ酸配列の同一性は、例えば、BLAST、FASTA等の当業者に公知のホモロジー検索プログラムを用いて算出することができる。CD3陰性かつCD56陽性である細胞の増殖活性を有するポリペプチドとは、本明細書の実施例に記載される細胞の培養方法に用いられる場合に、CD3陰性かつCD56陽性である細胞を含む細胞集団の細胞数を少なくとも2倍以上、好ましくは3倍以上に増幅させることができるポリペプチドをいう。 In this specification, the term 4-1BB ligand (4-1BBL) refers to a polypeptide or protein that can bind to 4-1BB (excluding antibodies). 4-1BBL may be synthesized by a method known to those skilled in the art, or may be artificially produced by recombinant gene technology, and 4-1BBL artificially produced by recombinant gene technology is called recombinant 4-1BBL. In one embodiment, 4-1BBL may be a polypeptide consisting of the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2 shown below, a polypeptide containing the extracellular domain of 4-1BBL (SEQ ID NO: 3), or a polypeptide consisting of an amino acid sequence having at least 70% or more, 80% or more, 85% or more, 90% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more identity with SEQ ID NO: 1, SEQ ID NO: 2, or SEQ ID NO: 3, and having proliferation activity of CD3-negative and CD56-positive cells. Amino acid sequence identity refers to the identity of the amino acid sequences between the target polypeptides, and is expressed as the percentage (%) of amino acid residues that match in the optimal alignment of the amino acid sequences. Amino acid sequence identity can be calculated using, for example, a homology search program known to those skilled in the art, such as BLAST or FASTA. A polypeptide having proliferation activity of CD3-negative and CD56-positive cells refers to a polypeptide that, when used in the cell culture method described in the Examples of this specification, can amplify the number of cells in a cell population containing CD3-negative and CD56-positive cells by at least two-fold, preferably three-fold.
(配列番号:1)
(SEQ ID NO: 1)
(配列番号:2)
(SEQ ID NO:2)
(配列番号:3)
(SEQ ID NO:3)
4−1BBLは、単量体であっても多量体であってもよい。多量体とは、複数のサブユニットが分子間相互作用、非共有結合、共有結合等により会合したものをいう。一つの態様において、本発明の4−1BBアゴニストは、ウレルマブ、ウトミルマブ、および4−1BBリガンドからなる群より選択される。本発明の製造方法の好ましい態様においては、4−1BBアゴニストは、ウレルマブ、および配列番号1と90%以上の同一性を有するアミノ酸配列からなり、かつCD3陰性かつCD56陽性である細胞の増殖活性を有するポリペプチドから選択される1以上である。なお、使用するサイトカイン、ポリペプチド等は、ヒトのアミノ酸配列を有することが好ましい。 4-1BBL may be a monomer or a multimer. A multimer refers to a combination of multiple subunits through intermolecular interactions, non-covalent bonds, covalent bonds, or the like. In one embodiment, the 4-1BB agonist of the present invention is selected from the group consisting of urelumab, utomirumab, and a 4-1BB ligand. In a preferred embodiment of the manufacturing method of the present invention, the 4-1BB agonist is one or more selected from urelumab and a polypeptide having an amino acid sequence with 90% or more identity to SEQ ID NO: 1 and having proliferation activity of CD3-negative and CD56-positive cells. It is preferable that the cytokine, polypeptide, etc. used have a human amino acid sequence.
好ましい態様においては、本発明の細胞集団の製造方法の工程a)における培地は、IL−2、IL−21および4−1BBアゴニストを含む。 In a preferred embodiment, the medium in step a) of the method for producing a cell population of the present invention contains IL-2, IL-21 and a 4-1BB agonist.
本発明の培地中のIL−2の濃度は、特に限定されず、50IU/mL以上とすることができ、100IU/mL以上であることが好ましく、150IU/mL以上であることがより好ましく、200IU/mL以上であることがさらに好ましく、250IU/mL以上であることが特に好ましく、281IU/mL以上であることが最も好ましい。また、工程a)の培地中のIL−2の濃度は、例えば、3,000IU/mL以下、2,810IU/mL以下、2,500IU/mL以下、2,000IU/mL以下、1,500IU/mL以下、1,000IU/mL以下、500IU/mL以下、または300IU/mL以下であってよい。前記したIL−2の濃度のいずれかの上限と下限とを組み合わせて、IL−2の濃度範囲として表してもよい。 The concentration of IL-2 in the medium of the present invention is not particularly limited, and can be 50 IU/mL or more, preferably 100 IU/mL or more, more preferably 150 IU/mL or more, even more preferably 200 IU/mL or more, particularly preferably 250 IU/mL or more, and most preferably 281 IU/mL or more. The concentration of IL-2 in the medium in step a) may be, for example, 3,000 IU/mL or less, 2,810 IU/mL or less, 2,500 IU/mL or less, 2,000 IU/mL or less, 1,500 IU/mL or less, 1,000 IU/mL or less, 500 IU/mL or less, or 300 IU/mL or less. The upper and lower limits of any of the above-mentioned IL-2 concentrations may be combined to represent the concentration range of IL-2.
本発明の製造方法の工程a)の培地中のIL−21の濃度は、特に限定されず、0.01ng/mL以上とすることができ、0.05ng/mL以上であることが好ましく、0.1ng/mL以上であることがより好ましく、0.5ng/mL以上であることがさらに好ましく、1ng/mL以上であることが特に好ましい。また、工程a)の培地中のIL−21の濃度は、例えば、1000ng/mL以下とすることができ、500ng/mL以下であってもよく、100ng/mL以下であることが好ましく、50ng/mL以下であることがより好ましく、10ng/mL以下であることがさらに好ましく、9ng/mL以下、8ng/mL以下、7ng/mL以下、6ng/mL以下、5ng/mL以下、4ng/mL以下、3ng/mL以下、または2ng/mL以下であってもよい。前記したIL−21の濃度のいずれかの上限と下限とを組み合わせて、IL−21の濃度範囲として表してもよい。 The concentration of IL-21 in the medium in step a) of the production method of the present invention is not particularly limited, and may be 0.01 ng/mL or more, preferably 0.05 ng/mL or more, more preferably 0.1 ng/mL or more, even more preferably 0.5 ng/mL or more, and particularly preferably 1 ng/mL or more. The concentration of IL-21 in the medium in step a) may be, for example, 1000 ng/mL or less, may be 500 ng/mL or less, preferably 100 ng/mL or less, more preferably 50 ng/mL or less, even more preferably 10 ng/mL or less, and may be 9 ng/mL or less, 8 ng/mL or less, 7 ng/mL or less, 6 ng/mL or less, 5 ng/mL or less, 4 ng/mL or less, 3 ng/mL or less, or 2 ng/mL or less. The upper and lower limits of any of the above-mentioned IL-21 concentrations may be combined to represent the concentration range of IL-21.
本発明の製造方法の工程a)の培地中の4−1BBアゴニストの濃度は、特に限定されず、0.01μg/mL以上とすることができ、0.05μg/mL以上であることが好ましく、0.1μg/mL以上であることがより好ましく、0.5μg/mL以上であることがさらに好ましく、1μg/mL以上であることが特に好ましい。また、工程a)の培地中の4−1BBアゴニストの濃度は、特に限定されず、例えば、1000μg/mL以下とすることができ、500μg/mL以下であることが好ましく、100μg/mL以下であることがより好ましく、50μg/mL以下であることがさらに好ましい。前記した4−1BBアゴニストの濃度のいずれかの上限と下限とを組み合わせて、4−1BBアゴニストの濃度範囲として表してもよい。 The concentration of the 4-1BB agonist in the medium in step a) of the production method of the present invention is not particularly limited, and can be 0.01 μg/mL or more, preferably 0.05 μg/mL or more, more preferably 0.1 μg/mL or more, even more preferably 0.5 μg/mL or more, and particularly preferably 1 μg/mL or more. The concentration of the 4-1BB agonist in the medium in step a) is not particularly limited, and can be, for example, 1000 μg/mL or less, preferably 500 μg/mL or less, more preferably 100 μg/mL or less, and even more preferably 50 μg/mL or less. The upper and lower limits of any of the above-mentioned concentrations of the 4-1BB agonist may be combined to express the concentration range of the 4-1BB agonist.
4−1BBLアゴニストとして、抗4−1BB抗体を用いる場合、本発明の製造方法の工程a)の培地中の抗4−1BB抗体の濃度は、0.01μg/mL以上とすることができ、0.05μg/mL以上であることが好ましく、0.1μg/mL以上であることがより好ましく、0.5μg/mL以上であることがさらに好ましく、1μg/mL以上であることが特に好ましい。また、工程a)の培地中の4−1BBLの濃度は、100μg/mL以下であることが好ましく、50μg/mL以下であることがより好ましく、10μg/mL以下であることがさらに好ましく、9μg/mL以下、8μg/mL以下、7μg/mL以下、6μg/mL以下、5μg/mL以下、4μg/mL以下、3μg/mL以下、または2μg/mL以下であってもよい。 When an anti-4-1BB antibody is used as the 4-1BBL agonist, the concentration of the anti-4-1BB antibody in the medium in step a) of the production method of the present invention can be 0.01 μg/mL or more, preferably 0.05 μg/mL or more, more preferably 0.1 μg/mL or more, even more preferably 0.5 μg/mL or more, and particularly preferably 1 μg/mL or more. In addition, the concentration of 4-1BBL in the medium in step a) is preferably 100 μg/mL or less, more preferably 50 μg/mL or less, even more preferably 10 μg/mL or less, and may be 9 μg/mL or less, 8 μg/mL or less, 7 μg/mL or less, 6 μg/mL or less, 5 μg/mL or less, 4 μg/mL or less, 3 μg/mL or less, or 2 μg/mL or less.
4−1BBLアゴニストとして4−1BBLを用いる場合、本発明の製造方法の工程a)の培地中の抗4−1BBLの濃度は、0.1μg/mL以上とすることができ、0.5μg/mL以上であることが好ましく、1μg/mL以上であることがより好ましく、2μg/mL以上、3μg/mL以上、4μg/mL以上、または5μg/mL以上であることがさらに好ましく、10μg/mL以上であることが特に好ましい。また、工程a)の培地中の4−1BBLの濃度は、例えば、1000μg/mL以下とすることができ、500μg/mL以下であることが好ましく、100μg/mL以下であることがより好ましく、50μg/mL以下であることがさらに好ましい。 When 4-1BBL is used as the 4-1BBL agonist, the concentration of anti-4-1BBL in the medium in step a) of the production method of the present invention can be 0.1 μg/mL or more, preferably 0.5 μg/mL or more, more preferably 1 μg/mL or more, even more preferably 2 μg/mL or more, 3 μg/mL or more, 4 μg/mL or more, or 5 μg/mL or more, and particularly preferably 10 μg/mL or more. In addition, the concentration of 4-1BBL in the medium in step a) can be, for example, 1000 μg/mL or less, preferably 500 μg/mL or less, more preferably 100 μg/mL or less, and even more preferably 50 μg/mL or less.
本発明の培地中において、IL−21および4−1BBアゴニストは液性因子ある。液性とは、目的のタンパク質、サイトカイン等の物質が固定化されることなく、培地において溶解可能であることをいい、液性因子とは、液性であるタンパク質等をいう。これに相反する概念としては、例えば、非特許文献4に記載されるような目的のタンパク質等がビーズに固定化された状態であることや、非特許文献5に記載されるようなfeeder細胞を形質転換し、feeder細胞の細胞膜上に目的のタンパク質等が発現された状態であること等が挙げられる。液性因子を含む培地は、好ましくは液体である。 In the medium of the present invention, IL-21 and 4-1BB agonists are humoral factors. "Hydrologic" means that the target protein, cytokine, or other substance is not immobilized and can dissolve in the medium, and humoral factors refer to proteins, etc. that are liquid. Opposite concepts include, for example, a state in which the target protein, etc. is immobilized on beads as described in Non-Patent Document 4, and a state in which the target protein, etc. is expressed on the cell membrane of the feeder cells after transformation as described in Non-Patent Document 5. The medium containing humoral factors is preferably liquid.
(培地)
本発明の培地は、特に限定されるものではないが、例えば、KBM501培地(KOHJIN BIO社)、KBM502培地(KOHJIN BIO社)、NTI培地(FUKOKU社)、コスメディウム008(コスモバイオ社)、FKCM101(FUKOKU社)、CellGro SCGM培地(セルジェニックス、岩井化学薬品株式会社)、X−VIVO15培地(ロンザ、タカラバイオ株式会社)、Gibco(登録商標) CTS(登録商標) AIM V(登録商標) Medium(サーモフィッシャーサイエンティフィック。T細胞および樹状細胞を増殖・操作するための既知組成の無血清培地)、CTS OpTmizer T Cell Expansion Basal Medium(サーモフィッシャーサイエンティフィック。ヒトTリンパ球の成長および増殖用)、IMDM、MEM、DMEM、RPMI−1640である。なお、本発明に関し、細胞について培養(する)というときは、特に記載した場合を除き、細胞の生存維持、細胞の増幅、および細胞の活性化からなる群より選択されるいずれかの目的のために細胞を一定時間、培地またはそれに準じた液の中で維持することをいう。処理を特定の温度で一定時間行う場合に、インキュベート(する)ということがある。
(Culture medium)
The medium of the present invention is not particularly limited, and examples thereof include KBM501 medium (KOHJIN BIO), KBM502 medium (KOHJIN BIO), NTI medium (FUKOKU), Cosmedium 008 (Cosmo Bio), FKCM101 (FUKOKU), CellGro SCGM medium (CellGenics, Iwai Chemicals Co., Ltd.), X-VIVO15 medium (Lonza, Takara Bio Inc.), Gibco (registered trademark) CTS (registered trademark) AIM V (registered trademark) Medium (Thermo Fisher Scientific, a chemically defined serum-free medium for growing and manipulating T cells and dendritic cells), CTS OpTmizer T Cell Expansion Basal Medium (Thermo Fisher Scientific, for growth and proliferation of human T lymphocytes), IMDM, MEM, DMEM, RPMI-1640. In the present invention, unless otherwise specified, the term "culturing" cells refers to maintaining cells in a medium or a liquid equivalent thereto for a certain period of time for any purpose selected from the group consisting of maintaining cell survival, expanding cells, and activating cells. "Incubating" may be used when a process is carried out at a specific temperature for a certain period of time.
培地は、IL−2を含むものであってもよい。IL−2を含む培地としては、KBM501培地(IL−2 2,810IU/mL(1,750JRU/mL)含有)、KBM502培地(IL−2 281IU/mL(175JRU/mL)含有)、コスメディウム008培地(IL−2 1,750JRU/mL含有)、FKCM101−L300培地(FUKOKU社、IL−2 300IU/mL含有)、FKCM101−L13T培地(FUKOKU社、IL−2 1,300IU/mL含有)等が挙げられる。本発明の製造方法の好ましい態様においては、培地は、KBM502培地またはKBM501培地を含んでいてもよい。 The medium may contain IL-2. Examples of media containing IL-2 include KBM501 medium (containing 2,810 IU/mL (1,750 JRU/mL) of IL-2), KBM502 medium (containing 281 IU/mL (175 JRU/mL) of IL-2), Cosmedium 008 medium (containing 1,750 JRU/mL of IL-2), FKCM101-L300 medium (FUKOKU, containing 300 IU/mL of IL-2), and FKCM101-L13T medium (FUKOKU, containing 1,300 IU/mL of IL-2). In a preferred embodiment of the production method of the present invention, the medium may contain KBM502 medium or KBM501 medium.
培地の交換または補充は、目的とする培養効果が得られることを条件として、培養開始後どの時点において行われてもかまわないが、2~5日毎が好ましい。 The medium may be replaced or replenished at any time after the start of culture, provided that the desired culture effect is obtained, but it is preferable to do so every 2 to 5 days.
培養に用いる培養容器は、商業的に入手可能なディッシュ、フラスコ、プレート、マルチウェルプレートを含むが、これらに限定されない。培養容器の他に、支持体等の様々な培養材料を用いて培養を行ってもよい。培養条件は、培養効果を損なわないことを条件として特に限定されないが、37℃、5%CO2および飽和水蒸気雰囲気下の培養条件が一般的である。 The culture vessels used for the culture include, but are not limited to, commercially available dishes, flasks, plates, and multi-well plates. In addition to the culture vessels, various culture materials such as supports may be used for the culture. The culture conditions are not particularly limited as long as they do not impair the culture effect, but the culture conditions are generally 37°C, 5% CO2, and saturated water vapor atmosphere.
(添加物等)
本発明の培地は、ヒト血清アルブミンを含んでいてもよい。ヒト血清アルブミンを培地に添加する場合には、例えば、日本赤十字社等から入手可能な5~25%ヒト血清アルブミン製剤を用いることができる。培地中のヒト血清アルブミンの濃度は、2,000mg/L以下であることが好ましい。
本発明の培地は、抗生物質を含んでいてもよい。抗生物質としては、例えば、硫酸カナマイシンを用いることができる。硫酸カナマイシンを培地に添加する場合には、例えば、Meiji Seika ファルマ社から入手可能な硫酸カナマイシン注射液を用いることができる。培地中の硫酸カナマイシンの濃度は、60mg/L以下であることが好ましい。
本発明の培地は、さらにヒトトランスフェリン、組換え型ヒトインスリン等を含むものであってもよい。
(Additives, etc.)
The medium of the present invention may contain human serum albumin. When human serum albumin is added to the medium, for example, a 5 to 25% human serum albumin preparation available from the Japanese Red Cross Society or the like can be used. The concentration of human serum albumin in the medium is preferably 2,000 mg/L or less.
The medium of the present invention may contain an antibiotic. For example, kanamycin sulfate can be used as the antibiotic. When kanamycin sulfate is added to the medium, for example, kanamycin sulfate injection available from Meiji Seika Pharma can be used. The concentration of kanamycin sulfate in the medium is preferably 60 mg/L or less.
The medium of the present invention may further contain human transferrin, recombinant human insulin, and the like.
本発明の培地は、IL−18をさらに含んでいてもよい。この場合、培地中のIL−18の濃度は、10~1000ng/mLであることが好ましく、100ng/mLであることがより好ましい。 The medium of the present invention may further contain IL-18. In this case, the concentration of IL-18 in the medium is preferably 10 to 1000 ng/mL, and more preferably 100 ng/mL.
培地には、本発明による課題の解決が阻害されない程度の範囲において、さらに、適切なタンパク質、サイトカイン、抗体、化合物その他の成分が含まれていてもよい。サイトカインは、上述したIL−2およびIL−21のほか、IL−3、IL−7、IL−12、IL−15、およびIL−18、幹細胞因子(SCF)、および/または、FMS様チロシンキナーゼ3リガンド(Flt3L)であり得る。これらはいずれも、ヒトのアミノ酸配列を有することが好ましく、安全上、組換えDNA技術で生産されることが好ましい。 The medium may further contain appropriate proteins, cytokines, antibodies, compounds and other components to the extent that the solution of the problems of the present invention is not hindered. In addition to the above-mentioned IL-2 and IL-21, the cytokines may be IL-3, IL-7, IL-12, IL-15, IL-18, stem cell factor (SCF), and/or FMS-like tyrosine kinase 3 ligand (Flt3L). All of these preferably have human amino acid sequences and are preferably produced by recombinant DNA technology for safety reasons.
本発明の培地は、1種以上の適切な添加物を含んでいてもよい。適切な添加物としては、例えば、ヒト血小板溶解物(HPL:Human Platelet Lysate)、ヒト血清代替物、ウシ胎児血清(FBS:Fetal Bovine Serum)等が挙げられる。本発明の培地が含む添加物としては、HPLまたはFBSが好ましく、HPLが特に好ましい。HPLとしては、例えば、UltraGRO(AventaCel社)を用いることができる。培地中のHPLの濃度は、1~10%であることが好ましく、3~8%であることがより好ましく、5%であることが特に好ましい。HPLを用いる場合には、培地にヘパリンをさらに添加することが好ましい。ヘパリンとしては、ヘパリンナトリウム、ヘパリンカルシウム等を用いることができる。培地中のヘパリンの濃度は、1~5U/mLであることが好ましく、2U/mLであることが特に好ましい。FBSとしては、例えば、Nichirei社から入手可能なFBS製剤を用いることができる。培地中のFBSの濃度は、1~10%であることが好ましく。3~8%であることがより好ましく、5%であることが特に好ましい。ヒト血清代替物としては、例えば、CTS Immune Cell SR(gibco社)を用いることができる。培地中のヒト血清代替物の濃度は、1~10%であることが好ましく、3~8%であることがより好ましく、5%であることが特に好ましい。 The medium of the present invention may contain one or more suitable additives. Suitable additives include, for example, human platelet lysate (HPL), human serum substitute, fetal bovine serum (FBS), etc. The additives contained in the medium of the present invention are preferably HPL or FBS, and particularly preferably HPL. As HPL, for example, UltraGRO (AventaCel) can be used. The concentration of HPL in the medium is preferably 1 to 10%, more preferably 3 to 8%, and particularly preferably 5%. When HPL is used, it is preferable to further add heparin to the medium. As heparin, sodium heparin, calcium heparin, etc. can be used. The concentration of heparin in the medium is preferably 1 to 5 U/mL, and particularly preferably 2 U/mL. As the FBS, for example, an FBS preparation available from Nichirei can be used. The concentration of FBS in the medium is preferably 1 to 10%, more preferably 3 to 8%, and particularly preferably 5%. As the human serum substitute, for example, CTS Immune Cell SR (Gibco) can be used. The concentration of the human serum substitute in the medium is preferably 1 to 10%, more preferably 3 to 8%, and particularly preferably 5%.
本発明の培地は、1種以上の免疫抑制剤を含んでいてもよい。培地が免疫抑制剤を含むことにより、CD3陽性細胞の増殖が抑制され得る。免疫抑制剤は、例えば、タクロリムス、シクロスポリン等のカルシニューリン阻害剤、バシリキシマブ等のサイトカイン阻害剤等であり、好ましくは、タクロリムスおよびバシリキシマブから選択される1種以上である。培地中のタクロリムスの濃度は、例えば、0.01~100ng/mLであってよく、好ましくは0.1~50ng/mLであり、より好ましくは1~10ng/mLである。培地中のバシリキマブの濃度は、例えば、0.005~50μg/mLであってよく、好ましくは0.05~30μg/mLであり、より好ましくは0.5~10μg/mLである。タクロリムスは、例えば、アステラス製薬から入手可能なプログラフ注射液を用いることができる。バシリキシマブは、例えば、ノバルティスファーマ社から入手可能なシムレクト静注用を用いることができる。 The medium of the present invention may contain one or more immunosuppressants. When the medium contains an immunosuppressant, the proliferation of CD3 positive cells can be suppressed. The immunosuppressant is, for example, a calcineurin inhibitor such as tacrolimus or cyclosporine, or a cytokine inhibitor such as basiliximab, and is preferably one or more selected from tacrolimus and basiliximab. The concentration of tacrolimus in the medium may be, for example, 0.01 to 100 ng/mL, preferably 0.1 to 50 ng/mL, and more preferably 1 to 10 ng/mL. The concentration of basiliximab in the medium may be, for example, 0.005 to 50 μg/mL, preferably 0.05 to 30 μg/mL, and more preferably 0.5 to 10 μg/mL. As the tacrolimus, for example, Prograf injection available from Astellas Pharma can be used. Basiliximab can be, for example, Simulect for intravenous injection, available from Novartis Pharma.
好ましい態様において、本発明の培養方法は、feeder細胞を用いない。本明細書において、feeder細胞とは、目的とする細胞(本発明によって得られる細胞)とは別の細胞であって、目的の細胞の培養条件を整えるために、例えば、成長因子となるサイトカインの生成等の補助的役割を果たす細胞をいう。feeder細胞は、本発明の属する技術分野にて通常使用されるものを指し、例えば、遺伝子が導入された動物細胞株、各種サイトカイン等で処理された白血球細胞、T細胞、B細胞、単球、抗原提示細胞、マクロファージ等であり得る。feeder細胞の具体例としては、非特許文献5に記載されるような、膜結合型IL−21を発現する遺伝子組換え抗原提示細胞が挙げられる。feeder細胞は、ガンマ線照射や抗生物質による処理等により増殖能がほとんど失われた細胞であり得るが、増殖能が低下した、ないし失われた細胞に限定されるものではない。 In a preferred embodiment, the culture method of the present invention does not use feeder cells. In this specification, feeder cells refer to cells that are different from the target cells (cells obtained by the present invention) and play an auxiliary role, for example, by producing cytokines that serve as growth factors, in order to prepare the culture conditions for the target cells. Feeder cells refer to cells that are commonly used in the technical field to which the present invention belongs, and can be, for example, animal cell lines into which genes have been introduced, white blood cells treated with various cytokines, T cells, B cells, monocytes, antigen-presenting cells, macrophages, etc. Specific examples of feeder cells include genetically modified antigen-presenting cells that express membrane-bound IL-21, as described in Non-Patent Document 5. Feeder cells can be cells that have almost completely lost their proliferation ability due to gamma irradiation, treatment with antibiotics, etc., but are not limited to cells that have reduced or lost their proliferation ability.
(培養期間)
工程a)は、単核球を含む細胞集団からCD3陽性細胞およびCD34陽性細胞を除去した細胞集団を、所定の期間培養することを含み得る。所定の期間は、1日以上であり、または2日以上、3日以上、4日以上、5日以上、6日以上、7日以上、8日以上、9日以上、10日以上、11日以上、12日以上、13日以上、14日以上もしくはそれ以上であってよく、さらに30日以下、25日以下、20日以下、15日以下もしくはそれ以下であってよく、特に限定されない。工程a)における培養期間は、例えば1~20日間であってよく、好ましくは3~18日間であり、4~16日間であってもよく、5~15日間であってもよく、6~14日間であってもよい。期間中、同じ培地に添加してもよく、培地を交換してもよく(この場合も、培地が要件を満たす限り工程a)が継続されているといえる。)、播種時の細胞密度を調整すれば、培養期間中のメンテナンス(培地の添加、交換等)を行わないことが可能である。
(Culture period)
Step a) may include culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells for a predetermined period of time. The predetermined period may be 1 day or more, or 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, 11 days or more, 12 days or more, 13 days or more, 14 days or more, or more, and may be 30 days or less, 25 days or less, 20 days or less, 15 days or less, or less, and is not particularly limited. The culture period in step a) may be, for example, 1 to 20 days, preferably 3 to 18 days, 4 to 16 days, 5 to 15 days, or 6 to 14 days. During the period, the cells may be added to the same medium, or the medium may be replaced (in this case, it can be said that step a) is continued as long as the medium meets the requirements.), and if the cell density at the time of seeding is adjusted, it is possible to avoid maintenance (addition, replacement, etc. of the medium) during the culture period.
<工程b>
本発明による細胞集団の製造方法は、工程a)で培養した細胞集団を、所定の培地で継代し、培養する工程b)をさらに含んでいてもよい。
<Step b>
The method for producing a cell population according to the present invention may further comprise the step b) of subculturing the cell population cultured in step a) in a predetermined medium.
本明細書において、継代は、細胞集団を新たな培地に移すことをいい、その方法は特に限定されない。細胞集団を新たな培地に移すことは、例えば、細胞を分散させた培地の一部を採取し新たな培地に加えることであってよい。継代する際に、継代前の培養に用いた培地の一部が新たな培地に加えられる場合があるが、本明細書においては、継代後の新たな培地中の組成においては、継代前の培養に用いた培地中の組成を無視することができる。 In this specification, passaging refers to transferring a cell population to a new medium, and the method is not particularly limited. Transferring a cell population to a new medium may involve, for example, collecting a portion of the medium in which the cells are dispersed and adding it to the new medium. When passaging, a portion of the medium used for the culture before passaging may be added to the new medium, but in this specification, the composition of the medium used for the culture before passaging can be ignored in the composition of the new medium after passaging.
本発明の細胞集団の製造方法の工程b)における培地は、IL−2を含む。工程b)における培地は、4−1BBアゴニストをさらに含んでいてもよく、IL−21をさらに含んでいてもよい。IL−2、4−1BBアゴニストおよびIL−21の具体的な説明に関しては、前述した内容を参照することができる。好ましい態様においては、本発明の細胞集団の製造方法の工程b)における培地は、IL−2および4−1BBアゴニストを含み、IL−21を含まない。 The medium in step b) of the method for producing a cell population of the present invention contains IL-2. The medium in step b) may further contain a 4-1BB agonist, and may further contain IL-21. For a specific description of IL-2, 4-1BB agonist, and IL-21, please refer to the above. In a preferred embodiment, the medium in step b) of the method for producing a cell population of the present invention contains IL-2 and a 4-1BB agonist, but does not contain IL-21.
工程b)の培地がIL−21を含む場合、培地中のIL−21の濃度は、例えば、0.01ng/mL以上、0.1ng/mL以上、または1ng/mL以上であってよく、一方で、100ng/mL以下であることが好ましく、50ng/mL以下であることがより好ましく、10ng/mL以下であることがさらに好ましく、9μg/mL以下、8μg/mL以下、7μg/mL以下、6μg/mL以下、5μg/mL以下、4μg/mL以下、3μg/mL以下、または2μg/mL以下であってもよい。 When the medium in step b) contains IL-21, the concentration of IL-21 in the medium may be, for example, 0.01 ng/mL or more, 0.1 ng/mL or more, or 1 ng/mL or more, while it is preferably 100 ng/mL or less, more preferably 50 ng/mL or less, even more preferably 10 ng/mL or less, and may be 9 μg/mL or less, 8 μg/mL or less, 7 μg/mL or less, 6 μg/mL or less, 5 μg/mL or less, 4 μg/mL or less, 3 μg/mL or less, or 2 μg/mL or less.
工程b)の培地が4−1BBアゴニストを含む場合、培地中の4−1BBアゴニストの濃度は、例えば、0.01μg/mL以上、0.1μg/mL以上、1μg/mL以上、5μg/mL以上、10μg/mL以上、または100μg/mL以上とすることができる。工程b)の培地中の4−1BBアゴニストの濃度は、好ましくは、工程a)の培地中の4−1BBアゴニストの濃度以上である。 When the medium in step b) contains a 4-1BB agonist, the concentration of the 4-1BB agonist in the medium can be, for example, 0.01 μg/mL or more, 0.1 μg/mL or more, 1 μg/mL or more, 5 μg/mL or more, 10 μg/mL or more, or 100 μg/mL or more. The concentration of the 4-1BB agonist in the medium in step b) is preferably equal to or greater than the concentration of the 4-1BB agonist in the medium in step a).
4−1BBLアゴニストとして抗4−1BB抗体を用いる場合、工程b)の培地中の抗4−1BB抗体の濃度は、0.01μg/mL以上とすることができ、0.05μg/mL以上であることが好ましく、0.1μg/mL以上であることがより好ましく、0.5μg/mL以上であることがさらに好ましく、1μg/mL以上であることが特に好ましい。4−1BBLアゴニストとして抗4−1BB抗体を用いる場合、工程b)の培地中の抗4−1BB抗体の濃度は、工程a)の培地中の抗4−1BB抗体の濃度以上であることが好ましい。 When an anti-4-1BB antibody is used as the 4-1BBL agonist, the concentration of the anti-4-1BB antibody in the medium in step b) can be 0.01 μg/mL or more, preferably 0.05 μg/mL or more, more preferably 0.1 μg/mL or more, even more preferably 0.5 μg/mL or more, and particularly preferably 1 μg/mL or more. When an anti-4-1BB antibody is used as the 4-1BBL agonist, the concentration of the anti-4-1BB antibody in the medium in step b) is preferably equal to or higher than the concentration of the anti-4-1BB antibody in the medium in step a).
4−1BBLアゴニストとして4−1BBLを用いる場合、工程b)の培地中の4−1BBLの濃度は、0.1μg/mL以上とすることができ、0.5μg/mL以上であることが好ましく、1μg/mL以上であることがより好ましく、5μg/mL以上であることがさらに好ましく、10μg/mL以上であることが特に好ましい。4−1BBLアゴニストとして4−1BBLを用いる場合、工程b)の培地中の4−1BBLの濃度は、工程a)の培地中の4−1BBLの濃度以上であることが好ましい。 When 4-1BBL is used as the 4-1BBL agonist, the concentration of 4-1BBL in the medium in step b) can be 0.1 μg/mL or more, preferably 0.5 μg/mL or more, more preferably 1 μg/mL or more, even more preferably 5 μg/mL or more, and particularly preferably 10 μg/mL or more. When 4-1BBL is used as the 4-1BBL agonist, the concentration of 4-1BBL in the medium in step b) is preferably equal to or greater than the concentration of 4-1BBL in the medium in step a).
工程b)は、工程a)で培養した細胞集団を、所定の期間培養することを含み得る。所定の期間は、1日以上であり、または2日以上、3日以上、4日以上、5日以上、6日以上、7日以上、8日以上、9日以上、10日以上、11日以上、12日以上、13日以上、14日以上、15日以上もしくはそれ以上であってよく、さらに30日以下、25日以下、20日以下もしくはそれ以下であってよく、特に限定されない。工程b)における培養期間は、例えば1~日間であってよく、好ましくは2~21日間であり、より好ましくは3~19日間であり、さらに好ましくは4~17日間であり、特に好ましくは8~15日間である。期間中、同じ培地に添加してもよく、培地を交換してもよい(この場合も、培地が要件を満たす限り工程b)が継続されているといえる。)。 Step b) may include culturing the cell population cultured in step a) for a predetermined period of time. The predetermined period may be 1 day or more, or 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, 11 days or more, 12 days or more, 13 days or more, 14 days or more, 15 days or more, or more, and may be 30 days or less, 25 days or less, 20 days or less, or less, and is not particularly limited. The culture period in step b) may be, for example, 1 to 2 days, preferably 2 to 21 days, more preferably 3 to 19 days, even more preferably 4 to 17 days, and particularly preferably 8 to 15 days. During the period, the cells may be added to the same medium, or the medium may be replaced (in this case, it can be said that step b) is continued as long as the medium meets the requirements).
<工程c>
本発明による細胞集団の製造方法は、工程b)で培養した細胞集団を、所定の培地で継代し、培養する工程c)を含んでいてもよい。
<Step c>
The method for producing a cell population according to the present invention may include a step c) of subculturing and culturing the cell population cultured in step b) in a predetermined medium.
本発明の細胞集団の製造方法の工程c)における培地は、IL−2を含む。工程c)における培地中は、4−1BBアゴニストをさらに含んでいてもよく、IL−21をさらに含んでいてもよい。IL−2、4−1BBアゴニストおよびIL−21の具体的な説明に関しては、前述した内容を参照することができる。一つの好ましい態様においては、本発明の工程c)における培地は、IL−2を含み、IL−21および4−1BBアゴニストを含まない。一つのより好ましい態様においては、本発明の工程c)における培地は、IL−2、IL−21および4−1BBアゴニストを含む。 The medium in step c) of the method for producing a cell population of the present invention contains IL-2. The medium in step c) may further contain a 4-1BB agonist, or may further contain IL-21. For a specific description of IL-2, 4-1BB agonist, and IL-21, please refer to the above. In one preferred embodiment, the medium in step c) of the present invention contains IL-2, but does not contain IL-21 or a 4-1BB agonist. In one more preferred embodiment, the medium in step c) of the present invention contains IL-2, IL-21, and a 4-1BB agonist.
工程c)の培地がIL−21を含む場合、培地中のIL−21の濃度は、例えば、0.01ng/mL以上、0.1ng/mL以上、もしくは1ng/mL以上、または100ng/mL以下、10ng/mL以下、5ng/mL以下とすることができる。 When the medium in step c) contains IL-21, the concentration of IL-21 in the medium can be, for example, 0.01 ng/mL or more, 0.1 ng/mL or more, or 1 ng/mL or more, or 100 ng/mL or less, 10 ng/mL or less, or 5 ng/mL or less.
工程c)の培地が4−1BBアゴニストを含む場合、培地中の4−1BBアゴニストの濃度は、例えば、0.01μg/mL以上、0.1μg/mL以上、もしくは1μg/mL以上、または100μg/mL以下、10μg/mL以下、5μg/mL以下とすることができる。 When the medium in step c) contains a 4-1BB agonist, the concentration of the 4-1BB agonist in the medium can be, for example, 0.01 μg/mL or more, 0.1 μg/mL or more, or 1 μg/mL or more, or 100 μg/mL or less, 10 μg/mL or less, or 5 μg/mL or less.
4−1BBLアゴニストとして抗4−1BB抗体を用いる場合、工程c)の培地中の抗4−1BB抗体の濃度は、0.01μg/mL以上とすることができ、0.05μg/mL以上であることが好ましく、0.1μg/mL以上であることがより好ましく、0.5μg/mL以上であることがさらに好ましく、1μg/mL以上であることが特に好ましく、または100μg/mL以下、10μg/mL以下、5μg/mL以下、4μg/mL以下、3μg/mL以下、2μg/mL以下であってもよい。 When an anti-4-1BB antibody is used as the 4-1BBL agonist, the concentration of the anti-4-1BB antibody in the medium in step c) can be 0.01 μg/mL or more, preferably 0.05 μg/mL or more, more preferably 0.1 μg/mL or more, even more preferably 0.5 μg/mL or more, and particularly preferably 1 μg/mL or more, or may be 100 μg/mL or less, 10 μg/mL or less, 5 μg/mL or less, 4 μg/mL or less, 3 μg/mL or less, or 2 μg/mL or less.
4−1BBLアゴニストとして4−1BBLを用いる場合、工程c)の培地中の4−1BBLの濃度は、0.1μg/mL以上とすることができ、0.5μg/mL以上であることが好ましく、1μg/mL以上であることがより好ましく、5μg/mL以上であることがさらに好ましく、10μg/mL以上であることが特に好ましい。 When 4-1BBL is used as the 4-1BBL agonist, the concentration of 4-1BBL in the medium in step c) can be 0.1 μg/mL or more, preferably 0.5 μg/mL or more, more preferably 1 μg/mL or more, even more preferably 5 μg/mL or more, and particularly preferably 10 μg/mL or more.
工程c)は、工程b)で培養した細胞集団を、所定の期間培養することを含み得る。所定の期間は、1日以上であり、2日以上、3日以上、4日以上、5日以上、6日以上、7日以上、8日以上、9日以上、10日以上もしくはそれ以上であってよく、さらに30日以下、25日以下、20日以下、15日以下、14日以下、13日以下、12日以下、11日以下もしくはそれ以下であってよく、特に限定されない。工程c)における培養期間は、好ましくは4~10日間であり、より好ましくは5~9日間であり、さらに好ましくは6~8日間であり、特に好ましくは7日である。期間中、同じ培地に添加してもよく、培地を交換してもよい(この場合も、培地が要件を満たす限り工程c)が継続されているといえる。)。 Step c) may include culturing the cell population cultured in step b) for a predetermined period of time. The predetermined period is 1 day or more, and may be 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, or 30 days or less, 25 days or less, 20 days or less, 15 days or less, 14 days or less, 13 days or less, 12 days or less, 11 days or less, or less, and is not particularly limited. The culture period in step c) is preferably 4 to 10 days, more preferably 5 to 9 days, even more preferably 6 to 8 days, and particularly preferably 7 days. During the period, the cells may be added to the same medium, or the medium may be replaced (in this case, it can be said that step c) is continued as long as the medium meets the requirements).
本発明による細胞集団の製造方法は、工程c)で培養した細胞集団を、工程c)と同様の条件でさらに培養する工程、すなわち工程c)を繰り返すことを含んでいてもよい。工程c)は、細胞の増殖が得られる限り、複数回繰り返してもよく、その回数は限定されない。 The method for producing a cell population according to the present invention may include a step of further culturing the cell population cultured in step c) under the same conditions as in step c), i.e., repeating step c). Step c) may be repeated multiple times, with no limit on the number of times, as long as cell proliferation is achieved.
本発明の製造方法により、工程a)に用いる単核球を含む細胞集団からCD3陽性細胞およびCD34陽性細胞を除去した細胞集団に含まれる細胞数は、少なくとも2倍以上に増幅され、好ましくは10倍以上に増幅され、より好ましくは100倍以上に増幅される。 By the manufacturing method of the present invention, the number of cells contained in the cell population obtained by removing CD3-positive cells and CD34-positive cells from the cell population containing mononuclear cells used in step a) is amplified at least 2-fold, preferably 10-fold, and more preferably 100-fold.
[本発明の製造方法によって得られる細胞集団]
本発明の製造方法によって得られる細胞集団は、少なくともCD3陰性かつCD56陽性であり、好ましくは、CD3陰性、CD34陰性、かつ、CCR5陽性、CCR6陽性、CXCR3陽性、ITGA1(Integrin α1)陽性およびITGA3(Integrin α3)陽性からなる群から選択される1以上からなる細胞表面マーカーの表現型を有する細胞を含む。より好ましくは、本発明の製造方法によって得られる細胞集団は、CD3陰性、CD34陰性、CCR5陽性、CCR6陽性、CXCR3陽性、ITGA1陽性かつITGA3陽性である細胞を含む。細胞表面マーカーとして用いるCCRまたはCXCRは、ケモカインレセプターの一種であり、ITG(Integrin)は、細胞接着因子の一種である。
[Cell population obtained by the production method of the present invention]
The cell population obtained by the production method of the present invention is at least CD3 negative and CD56 positive, and preferably contains cells having a phenotype of one or more cell surface markers selected from the group consisting of CD3 negative, CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 (Integrin α1) positive, and ITGA3 (Integrin α3) positive. More preferably, the cell population obtained by the production method of the present invention contains cells that are CD3 negative, CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, and ITGA3 positive. CCR or CXCR used as a cell surface marker is a type of chemokine receptor, and ITG (Integrin) is a type of cell adhesion factor.
本発明において、CD3陰性かつCD56陽性である細胞を含む細胞集団という場合、細胞集団中、CD3陰性かつCD56陽性である細胞の割合は、例えば、10%以上であってよく、20%以上であってよく、30%以上であってよく、40%以上であってよく、50%以上であってよく、好ましくは60%以上であり、より好ましくは70%以上であり、さらに好ましくは80%以上であり、特に好ましくは90%以上である。細胞集団中のCD3陰性かつCD56陽性である細胞の割合が前記したいずれかの割合より少ない細胞集団は、本発明によって得られるCD3陰性かつCD56陽性である細胞を含む細胞集団とは区別される。 In the present invention, when a cell population containing CD3-negative and CD56-positive cells is mentioned, the proportion of CD3-negative and CD56-positive cells in the cell population may be, for example, 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, preferably 60% or more, more preferably 70% or more, even more preferably 80% or more, and particularly preferably 90% or more. A cell population in which the proportion of CD3-negative and CD56-positive cells in the cell population is lower than any of the above proportions is distinguished from a cell population containing CD3-negative and CD56-positive cells obtained by the present invention.
また、本発明において、CD3陰性、CD56陽性であり、かつCD34陰性、CCR5陽性、CCR6陽性、CXCR3陽性、ITGA1陽性またはITGA3陽性である細胞を含む細胞集団という場合、細胞集団中、細胞表面マーカーの各表現型を有する細胞の割合は、例えば、10%以上であってよく、20%以上であってよく、30%以上であってよく、40%以上であってよく、50%以上であってよく、好ましくは60%以上であり、より好ましくは70%以上であり、さらに好ましくは80%以上であり、特に好ましくは90%以上であってよい。細胞集団中のCD3陰性かつCD56陽性であり、かつCD34陰性、CCR5陽性、CCR6陽性、CXCR3陽性、ITGA1陽性またはITGA3陽性である細胞の割合が前記したいずれかの割合より少ない細胞集団は、本発明によって得られるCD3陰性かつCD56陽性であり、かつCD34陰性、CCR5陽性、CCR6陽性、CXCR3陽性、ITGA1陽性またはITGA3陽性である細胞を含む細胞集団とは区別される。本発明によって得られる細胞集団中のCD3陰性かつCD56陽性である細胞は、好ましくは、CD34陰性、CCR5陽性、CCR6陽性、CXCR3陽性、ITGA1陽性かつITGA3陽性であり、より好ましくは、CD34陰性、CCR5陽性、CCR6陽性、CXCR3陽性、ITGA1陽性かつITGA3陽性である。 Furthermore, in the present invention, when a cell population is referred to that contains cells that are CD3 negative, CD56 positive, and CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, or ITGA3 positive, the percentage of cells having each phenotype of cell surface marker in the cell population may be, for example, 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, preferably 60% or more, more preferably 70% or more, even more preferably 80% or more, and particularly preferably 90% or more. A cell population in which the proportion of cells that are CD3 negative and CD56 positive, and CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, or ITGA3 positive is less than any of the above proportions, is distinguished from a cell population obtained by the present invention that contains cells that are CD3 negative and CD56 positive, and CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, or ITGA3 positive. The CD3 negative and CD56 positive cells in the cell population obtained by the present invention are preferably CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, and ITGA3 positive, and more preferably CD34 negative, CCR5 positive, CCR6 positive, CXCR3 positive, ITGA1 positive, and ITGA3 positive.
[細胞傷害活性]
本発明によって得られる細胞集団に含まれる細胞は、高い細胞傷害活性を発揮し得る。細胞傷害活性は、例えば、腫瘍細胞等の標的細胞(T)に対する、対象細胞(エフェクター細胞(E))の溶解能を測定することにより評価することができる。腫瘍細胞としては、例えば、白血病、グリオーマ、乳がん、大腸がん、卵巣がん、前立腺がん等の腫瘍細胞株を用いることができるが、これらに限定されない。また、標的細胞として、腫瘍細胞株から作製したスフェロイドを用いることもできる。エフェクター細胞と標的細胞の比(E:T)は、特に限定されず、例えば、1:1、2:1、5:1等であってよい。細胞傷害活性は、エフェクター細胞により死に至った標的細胞の百分率(%)で表すことができ、次式により求められる。
[Cytotoxic activity]
The cells contained in the cell population obtained by the present invention can exhibit high cytotoxic activity. The cytotoxic activity can be evaluated, for example, by measuring the lysis ability of target cells (effector cells (E)) against target cells (T) such as tumor cells. For example, tumor cell lines of leukemia, glioma, breast cancer, colon cancer, ovarian cancer, prostate cancer, etc. can be used as tumor cells, but are not limited thereto. In addition, spheroids prepared from tumor cell lines can also be used as target cells. The ratio of effector cells to target cells (E:T) is not particularly limited, and may be, for example, 1:1, 2:1, 5:1, etc. The cytotoxic activity can be expressed as the percentage (%) of target cells killed by effector cells, and is calculated by the following formula.
細胞傷害活性率(%Lysis)=(標的細胞死細胞率−陰性コントロール死細胞率(自然細胞死率))/(陽性コントロール死細胞率(最大細胞死率)−陰性コントロール死細胞率)×100 Cytotoxic activity rate (% Lysis) = (target cell death rate - negative control cell death rate (spontaneous cell death rate)) / (positive control cell death rate (maximum cell death rate) - negative control cell death rate) x 100
細胞傷害活性は、例えば、放射性物質、蛍光色素等で標識した抗体等の試薬により、エフェクター細胞と標的細胞、生細胞と死細胞を区別し、フローサイトメトリー法等を用いてこれらの細胞を定量して、上記式の各死細胞率を求めることによって測定することができる。本発明の細胞集団をエフェクター細胞として用いる場合、標的細胞とインキュベートする時間は、用いる標的細胞の種類によって適宜設定することができ、例えば、白血病細胞株であるK562細胞を標的細胞として用いる場合には、2時間とすることができる。 Cytotoxic activity can be measured, for example, by distinguishing between effector cells and target cells, and between live and dead cells, using reagents such as antibodies labeled with radioactive substances or fluorescent dyes, quantifying these cells using flow cytometry or the like, and determining the cell death rate according to the above formula. When the cell population of the present invention is used as effector cells, the incubation time with target cells can be set appropriately depending on the type of target cells used; for example, when K562 cells, a leukemia cell line, are used as target cells, it can be set to 2 hours.
本発明に関し、細胞傷害活性が高いというときは、特に記載した場合を除き、標的細胞をK562細胞とし、E:T=1~5:1で混合し、2時間共培養した場合の細胞傷害活性が50%以上であることをいう。細胞傷害活性は、60%以上であることが好ましく、70%以上であることがより好ましい。 In the present invention, unless otherwise specified, high cytotoxic activity means that the cytotoxic activity is 50% or more when the target cells are K562 cells, mixed at an E:T ratio of 1-5:1, and co-cultured for 2 hours. The cytotoxic activity is preferably 60% or more, and more preferably 70% or more.
また、本発明によって得られる細胞集団をエフェクター細胞として用いて細胞傷害活性を測定する場合、エフェクター細胞中のCD107a陽性細胞を検出することによっても、所望の細胞傷害活性が得られたと判断することができる。CD107aは、NK細胞内顆粒に存在し、脱顆粒されるとNK細胞の細胞膜表面上に移行することから、CD107a陽性であることは、間接的に、細胞傷害活性、すなわち本発明によって得られる細胞集団に含まれる細胞による標的細胞への攻撃が得られたことを示すためである。CD107a陽性細胞の検出は、例えば、蛍光標識した抗CD107a抗体を対象細胞とインキュベートさせた後の発光を測定することにより行うことができる。本発明によって得られる細胞集団をエフェクター細胞として、任意の腫瘍細胞株を標的細胞として、1~2:1の細胞数比(E:T)で用いた場合、本発明によって得られる細胞集団中のCD107a陽性細胞の割合は、少なくとも20%以上であり、好ましくは30%以上であり、より好ましくは40%以上であり、さらに好ましくは50%以上である。腫瘍細胞株は、例えば、白血病細胞株であるK562細胞等であってよい。 In addition, when measuring cytotoxic activity using the cell population obtained by the present invention as effector cells, the desired cytotoxic activity can also be determined by detecting CD107a-positive cells in the effector cells. Since CD107a is present in granules in NK cells and migrates to the cell membrane surface of NK cells upon degranulation, CD107a positivity indirectly indicates that cytotoxic activity, i.e., attack of target cells by cells contained in the cell population obtained by the present invention, has been obtained. CD107a-positive cells can be detected, for example, by measuring luminescence after incubating target cells with a fluorescently labeled anti-CD107a antibody. When the cell population obtained by the present invention is used as effector cells and any tumor cell line is used as target cells at a cell number ratio of 1 to 2:1 (E:T), the proportion of CD107a-positive cells in the cell population obtained by the present invention is at least 20% or more, preferably 30% or more, more preferably 40% or more, and even more preferably 50% or more. The tumor cell line may be, for example, a leukemia cell line, such as K562 cells.
[抗腫瘍効果]
本発明によって得られる細胞集団は、in vivoにて投与された場合においても、抗腫瘍効果を発揮する。in vivoでの抗腫瘍効果は、例えば、担がんモデル動物を用いて、本発明によって得られる細胞集団を投与し、腫瘍体積または腫瘍細胞数の変化を測定することにより評価することができる。担がんモデル動物の作製や腫瘍体積または腫瘍細胞数の変化の測定は、当業者に公知の方法を用いて行うことができるが、例えば、蛍光標識した腫瘍細胞を動物に移植し、観察する方法が挙げられる。具体例としては、ルシフェラーゼを発現させたSKOV細胞(ヒト卵巣がん細胞株)を免疫不全マウスに移植し、ルシフェリンをマウスに投与し、麻酔下で腫瘍の発光を観察するin vivoイメージングシステム(IVIS)を用いた方法等が挙げられる。この方法において、本発明によって得られる細胞集団の投与方法は特に限定されず、投与回数は単回または複数回であってよく、複数回投与する場合の投与間隔は適宜設定することができる。投与のタイミングは、例えば、腫瘍移植1日後~30日後の任意の時点とすることができる。投与用量は、例えば、1回あたり1.0×104~1.0×1010cells/bodyとすることができ、好ましくは1.0×105~1.0×109cells/bodyとすることができ、より好ましくは1.0×106~1.0×108cells/bodyとすることができ、特に好ましくは1.5×107~2.7×107cells/bodyとすることができる。投与経路は、任意であってよく、筋肉内、静脈内、動脈内、腹腔内または皮下投与であってよいが、好ましくは腹腔内投与が選択される。本発明によって得られる細胞集団の活性を生体内で維持する目的で、IL−2を同時に投与してもよい。この場合におけるIL−2の投与量は、1,000~10,000IU/bodyとすることができ、好ましくは5,000IU/bodyとすることができる。一つの態様において、本発明によって得られる細胞集団の投与は、投与しない場合と比較して、担がんモデル動物における腫瘍細胞数を1/10以下に減少させることができる。
[Antitumor effect]
The cell population obtained by the present invention exerts an antitumor effect even when administered in vivo. The antitumor effect in vivo can be evaluated, for example, by administering the cell population obtained by the present invention to a cancer-bearing model animal and measuring the change in tumor volume or the number of tumor cells. The preparation of the cancer-bearing model animal and the measurement of the change in tumor volume or the number of tumor cells can be performed using methods known to those skilled in the art, for example, a method of transplanting fluorescently labeled tumor cells into the animal and observing them. A specific example is a method using an in vivo imaging system (IVIS) in which luciferase-expressing SKOV cells (human ovarian cancer cell line) are transplanted into an immunodeficient mouse, luciferin is administered to the mouse, and the tumor luminescence is observed under anesthesia. In this method, the administration method of the cell population obtained by the present invention is not particularly limited, and the number of administrations may be single or multiple, and the administration interval when multiple administrations are performed can be appropriately set. The timing of administration can be any time point, for example, 1 to 30 days after tumor transplantation. The dosage may be, for example, 1.0×10 4 to 1.0×10 10 cells/body per administration, preferably 1.0×10 5 to 1.0×10 9 cells/body, more preferably 1.0×10 6 to 1.0×10 8 cells/body, and particularly preferably 1.5×10 7 to 2.7×10 7 cells/body. The route of administration may be any, including intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration, with intraperitoneal administration being preferred. IL-2 may be administered simultaneously in order to maintain the activity of the cell population obtained by the present invention in vivo. The dosage of IL-2 in this case may be 1,000 to 10,000 IU/body, and preferably 5,000 IU/body. In one embodiment, administration of the cell population obtained by the present invention can reduce the number of tumor cells in a cancer-bearing model animal to 1/10 or less, compared to the case where the cell population is not administered.
本発明によって得られる細胞集団は、凍結保存することが可能である。凍結保存は、例えば、ジメチルスルホキシド(DMSO)等の凍結保護試薬を含んだ完全培地等の凍結保存に適した培地を用いて、適切な細胞密度になるように細胞集団を懸濁し、クライオバイアル等の凍結保存用の容器に入れて、−80℃以下にて保管することにより行うことができる。適切な細胞密度は、例えば1×103~1×1010cells/mLであってよく、好ましくは2×108cells/5mLとすることができる。凍結保護試薬は、無機塩類やpH調節剤等を含んでいてもよい。 The cell population obtained by the present invention can be cryopreserved. Cryopreservation can be performed, for example, by suspending the cell population to an appropriate cell density using a medium suitable for cryopreservation, such as a complete medium containing a cryoprotective agent such as dimethyl sulfoxide (DMSO), placing the suspension in a cryopreservation container such as a cryovial, and storing the suspension at −80° C. or lower. The appropriate cell density may be, for example, 1×10 3 to 1×10 10 cells/mL, and preferably 2×10 8 cells/5 mL. The cryoprotective agent may contain inorganic salts, a pH adjuster, and the like.
さらに、凍結保存した細胞集団を適切な方法により解凍することで、凍結・解凍後も細胞集団の細胞傷害活性は維持され得る。適切な解凍の方法は、例えば、細胞集団の入った凍結保存用容器を37℃の水浴に移し、緩やかに旋回させることである。 Furthermore, by thawing the cryopreserved cell population using an appropriate method, the cytotoxic activity of the cell population can be maintained even after freezing and thawing. An appropriate thawing method is, for example, transferring the cryopreservation container containing the cell population to a 37°C water bath and gently swirling it.
[医薬組成物]
本発明の一つの態様によれば、本発明によって得られる細胞集団を含む医薬組成物が提供される。医薬組成物は、医薬として許容される添加物を含んでいてもよい。医薬として許容される添加物としては、例えば、例えば等張化剤、pH調整剤、緩衝剤、安定化剤、凍結保護剤、溶解補助剤、無痛化剤、抗生物質等が挙げられる。
Pharmaceutical Compositions
According to one aspect of the present invention, there is provided a pharmaceutical composition comprising the cell population obtained by the present invention. The pharmaceutical composition may contain a medicament acceptable additive. Examples of the medicament acceptable additive include an isotonicity agent, a pH adjuster, a buffer, a stabilizer, a cryoprotectant, a dissolution aid, a pain-relieving agent, and an antibiotic.
医薬組成物は、典型的には、本発明によって得られる細胞集団を溶液に懸濁した形態である。本発明によって得られる細胞集団を懸濁するための溶液は、例えば、DMSOを含む凍結用保護液、生理食塩水、リン酸緩衝生理食塩水(PBS)、培地、血清等が一般的である。溶液は、医薬品および医薬部外品として薬学的に許容される担体を含む場合がある。 The pharmaceutical composition is typically in the form of a suspension of the cell population obtained by the present invention in a solution. Typical solutions for suspending the cell population obtained by the present invention include, for example, cryoprotectants containing DMSO, saline, phosphate-buffered saline (PBS), culture medium, serum, etc. The solution may contain a pharma- ceutical acceptable carrier as a drug or quasi-drug.
本発明の医薬組成物は、感染症、または癌を治療するために用いられる場合がある。本発明の医薬組成物はまた、本発明の細胞集団に感受性を有するさまざまな疾患の治療に適用することができる。本発明の医薬組成物はまた、本発明の細胞集団に感受性を有するさまざまな疾患の予防に適用することができる。疾患は、例えば、口腔癌、胆嚢癌、胆管癌、肺癌、肝臓癌、大腸癌、腎臓癌、膀胱癌、白血病や、ウイルス、細菌等による感染症を含むが、これらに限定されない。本発明の細胞療法は、単独か、あるいは外科療法、化学療法、放射線療法、抗体医薬品等と組み合わせて実施される場合がある。本発明の医薬組成物を用いた細胞療法において、本発明の細胞集団は、例えば、静脈、動脈、皮下、腹腔内等へ投与される場合がある。 The pharmaceutical composition of the present invention may be used to treat infectious diseases or cancer. The pharmaceutical composition of the present invention may also be applied to the treatment of various diseases sensitive to the cell population of the present invention. The pharmaceutical composition of the present invention may also be applied to the prevention of various diseases sensitive to the cell population of the present invention. Diseases include, but are not limited to, oral cancer, gallbladder cancer, bile duct cancer, lung cancer, liver cancer, colon cancer, kidney cancer, bladder cancer, leukemia, and infectious diseases caused by viruses, bacteria, etc. The cell therapy of the present invention may be performed alone or in combination with surgical therapy, chemotherapy, radiation therapy, antibody drugs, etc. In cell therapy using the pharmaceutical composition of the present invention, the cell population of the present invention may be administered, for example, into a vein, an artery, subcutaneously, or intraperitoneally.
本発明の医薬組成物は、本発明の細胞集団と異なるHLA遺伝子型を有する患者に投与される場合がある。 The pharmaceutical composition of the present invention may be administered to a patient having an HLA genotype different from that of the cell population of the present invention.
本発明の医薬組成物は、IL−2製剤とともに投与することができる。IL−2製剤は、遺伝子組換え型であってもよく、テセロイキン(遺伝子組換え、製品名:イムネース(塩野義製薬))であることができる。 The pharmaceutical composition of the present invention can be administered together with an IL-2 preparation. The IL-2 preparation may be a recombinant type, such as teceleukin (recombinant, product name: Immunace (Shionogi Pharmaceuticals)).
本発明の医薬組成物は、抗体医薬品と併用することができる場合がある。本発明の医薬組成物と併用できる抗体の具体例としては、ibritumomabtiuxetan、iodine131、catumaxomab、blinatumomab、muromonab−CD3、abciximab、rituximab、basiliximab、infliximab、cetuximab、brentuximab、siltuximab、dinutuximab、obiltoxaximab、daclizumab、palivizumab、trastuzumab、gemtuzumab、alemtuzumab、omalizumab、efalizumab、bevacizumab、natalizumab、tocilizumab、ranibizumab、eculizumab、certolizumabpegol、mogamulizumab、pertuzumab、trastuzumab、obinutuzumab、vedolizumab、pembrolizuma、idarucizumab、mepolizumab、elotuzumab、daratumumab、ixekizumab、reslizumab、adalimumab、panitumumab、golimumab、ustekinumab、canakinumab、ofatumumab、denosumab、ipilimumab、belimumab、raxibacumab、ramucirumab、nivolumab、secukinumab、evolocumab、alirocumab、およびnecitumumabを挙げることができる。 The pharmaceutical composition of the present invention may be used in combination with an antibody drug. Specific examples of antibodies that may be used in combination with the pharmaceutical composition of the present invention include ibritumomabtiuxetan, iodine131, catumaxomab, blinatumomab, muromonab-CD3, abciximab, rituximab, basiliximab, infliximab, cetuximab, brentuximab, siltuximab, diclofenac, and ribacilaximab. nutuximab, obiltoxaximab, daclizumab, palivizumab, trastuzumab, gemtuzumab, alemtuzumab, omal izumab, efalizumab, bevacizumab, natalizumab, tocilizumab, ranibizumab, eculizumab, certolizum abpegol, mogamulizumab, pertuzumab, trastuzumab, obinutuzumab, vedolizumab, pembrolizuma, ida rucizumab, mepolizumab, elotuzumab, daratumumab, ixekizumab, reslizumab, adalimumab, panitumum Examples of such agents include ribacumab, golimumab, ustekinumab, canakinumab, ofatumumab, denosumab, ipilimumab, belimumab, raxibacumab, ramucirumab, nivolumab, secukinumab, evolocumab, alirocumab, and necitumumab.
本発明の医薬組成物と併用することができる抗体は、CD16に親和性の高いものであることが好ましい。また、本発明の医薬組成物において、抗体の少なくとも一部は、本発明の細胞集団に含まれる細胞に結合していてもよい場合がある。 Antibodies that can be used in combination with the pharmaceutical composition of the present invention preferably have high affinity for CD16. In addition, in the pharmaceutical composition of the present invention, at least a portion of the antibody may be bound to cells contained in the cell population of the present invention.
本発明の医薬組成物に含まれる細胞は、キメラ抗原受容体(CAR)を発現したものであってもよい。CARを発現させる方法は、当業者において公知のものに従ってよく、例えば、目的のCARをコードする遺伝子を含むベクターを用いて、本発明の細胞集団に含まれる細胞にこれらの遺伝子を導入する方法であってよい。CARは、例えば、任意の抗体由来の短鎖領域フラグメント(scFv)と細胞内シグナル伝達ドメインとを含む構造を有していてよい。 The cells contained in the pharmaceutical composition of the present invention may express a chimeric antigen receptor (CAR). The method for expressing the CAR may be according to a method known to those skilled in the art, for example, a method in which a vector containing a gene encoding the desired CAR is used to introduce the gene into cells contained in the cell population of the present invention. The CAR may have a structure containing, for example, a short chain fragment (scFv) derived from any antibody and an intracellular signaling domain.
本発明の医薬組成物の製造は、医薬品および医薬部外品の製造管理および品質管理規則に適合した条件(good manufacturing practice、GMP)および再生医療等製品の製造管理および品質管理の基準(Good Gene,Cellular,and Tissue−based Products Manufacturing Practice、GCTP)で実施されることが好ましい。 The pharmaceutical composition of the present invention is preferably manufactured under conditions that comply with the manufacturing and quality control regulations for drugs and quasi-drugs (good manufacturing practice, GMP) and the standards for manufacturing and quality control for regenerative medicine products (Good Gene, Cellular, and Tissue-based Products Manufacturing Practice, GCTP).
以下に実施例を挙げて本発明をさらに具体的に説明するが、本発明はこれにより限定して解釈されるものではない。 The present invention will be explained in more detail below with reference to examples, but the present invention should not be construed as being limited thereto.
[培養方法]
《PBMCの解凍》
凍結保存PBMC(Peripheral Blood Mononuclear Cells:末梢血単核細胞)を37℃のウォーターバスにて完全に解凍し、RPMI1640培地(10%FBSおよび1%ペニシリン−ストレプトマイシン混合溶液を添加)にて10倍に希釈した。500g、5分間遠心後、各条件下にて使用した。
[Culture method]
Thawing PBMC
Cryopreserved PBMCs (Peripheral Blood Mononuclear Cells) were completely thawed in a water bath at 37° C. and diluted 10-fold with RPMI 1640 medium (containing 10% FBS and 1% penicillin-streptomycin mixed solution). After centrifugation at 500 g for 5 minutes, the cells were used under each condition.
A).実施例1(ドナーMixA)
培養液と添加物とが異なる条件でNK細胞の培養試験を行った。
A). Example 1 (Donor Mix A)
NK cell culture tests were carried out under different conditions of culture medium and additives.
・Day0
解凍したPBMCより、EasySep Human NK Cell Enrichment Kit(STEMCELL Technologies,19055)を使用してNK細胞(CD3−/CD56+,≧80%)を単離した。単離したNK細胞を以下に示す18群の培地にて2×105cells/mLに調製し、12ウェルプレートに0.65mL播種し培養を開始した。
・Day 0
NK cells (CD3-/CD56+, ≧80%) were isolated from the thawed PBMCs using EasySep Human NK Cell Enrichment Kit (STEMCELL Technologies, 19055). The isolated NK cells were adjusted to 2×10 5 cells/mL in the following 18 groups of medium, and 0.65 mL was seeded on a 12-well plate to start culture.
(1)RPMI1640培地,5%CTS,IL−2[100IU/mL],IL−21[100ng/mL,Urelumab[10μg/mL]
(2)RPMI1640培地,5%UltraGRO,ヘパリンナトリウム[2U/mL],IL−2[100IU/mL],IL−21[100ng/mL],Urelumab[10μg/mL]
(3)RPMI1640培地,5%FBS,IL−2[100IU/mL],IL−21[100ng/mL],Urelumab[10μg/mL]
(4)RPMI1640培地,5%CTS,IL−2[100IU/mL]
(5)RPMI1640培地,5%UltraGRO,ヘパリンナトリウム[2U/mL],IL−2[100IU/mL]
(6)RPMI1640培地,5%FBS,IL−2[100IU/mL]
(7)KBM501培地,5%CTS,IL−21[100ng/mL],Urelumab[10μg/mL]
(8)KBM501培地,5%UltraGRO,ヘパリンナトリウム[2U/mL],IL−21[100ng/mL],Urelumab[10μg/mL]
(9)KBM501培地,5%FBS,IL−21[100ng/mL],Urelumab[10μg/mL]
(10)KBM501培地,5%CTS
(11)KBM501培地,5%UltraGRO,ヘパリンナトリウム[2U/mL]
(12)KBM501培地,5%FBS
(13)NTI培地,5%CTS,IL−21[100ng/mL],Urelumab[10μg/mL]
(14)NTI培地,5%UltraGRO,ヘパリンナトリウム[2U/mL],IL−21[100ng/mL],Urelumab[10μg/mL]
(15)NTI培地,5%FBS,IL−21[100ng/mL],Urelumab[10μg/mL]
(16)NTI培地,5%CTS
(17)NTI培地,5%UltraGRO,ヘパリンナトリウム[2U/mL]
(18)NTI培地,5%FBS
(1) RPMI1640 medium, 5% CTS, IL-2 [100 IU/mL], IL-21 [100 ng/mL, Urelumab [10 μg/mL]
(2) RPMI 1640 medium, 5% UltraGRO, sodium heparin [2 U/mL], IL-2 [100 IU/mL], IL-21 [100 ng/mL], Urelumab [10 μg/mL]
(3) RPMI 1640 medium, 5% FBS, IL-2 [100 IU/mL], IL-21 [100 ng/mL], Urelumab [10 μg/mL]
(4) RPMI 1640 medium, 5% CTS, IL-2 [100 IU/mL]
(5) RPMI 1640 medium, 5% UltraGRO, sodium heparin [2 U/mL], IL-2 [100 IU/mL]
(6) RPMI 1640 medium, 5% FBS, IL-2 [100 IU/mL]
(7) KBM501 medium, 5% CTS, IL-21 [100 ng/mL], Urelumab [10 μg/mL]
(8) KBM501 medium, 5% UltraGRO, sodium heparin [2 U/mL], IL-21 [100 ng/mL], Urelumab [10 μg/mL]
(9) KBM501 medium, 5% FBS, IL-21 [100 ng/mL], Urelumab [10 μg/mL]
(10) KBM501 medium, 5% CTS
(11) KBM501 medium, 5% UltraGRO, sodium heparin [2 U/mL]
(12) KBM501 medium, 5% FBS
(13) NTI medium, 5% CTS, IL-21 [100 ng/mL], Urelumab [10 μg/mL]
(14) NTI medium, 5% UltraGRO, sodium heparin [2 U/mL], IL-21 [100 ng/mL], Urelumab [10 μg/mL]
(15) NTI medium, 5% FBS, IL-21 [100 ng/mL], Urelumab [10 μg/mL]
(16) NTI medium, 5% CTS
(17) NTI medium, 5% UltraGRO, sodium heparin [2 U/mL]
(18) NTI medium, 5% FBS
・Day7
全群、ウェルをよくピペッティングし細胞数をカウントした。(8)、(14)は6ウェルプレートに0.5mLを分取し、以下に示す培地の1mLを添加した。
Day 7
For all groups, the wells were thoroughly pipetted and the number of cells was counted. For (8) and (14), 0.5 mL was dispensed into a 6-well plate, and 1 mL of the medium shown below was added.
(8)KBM501培地,5%UltraGRO,ヘパリンナトリウム[2U/mL]
(14)NTI培地,5%UltraGRO,ヘパリンナトリウム[2U/mL]
(8) KBM501 medium, 5% UltraGRO, sodium heparin [2 U/mL]
(14) NTI medium, 5% UltraGRO, sodium heparin [2 U/mL]
・Day11
全群、ウェルをよくピペッティングし細胞数をカウントした。
・Day 11
In all groups, the wells were thoroughly pipetted and the number of cells was counted.
結果を表1および図1に示す。 The results are shown in Table 1 and Figure 1.
RPMI培地、KBM培地またはNTI培地を用いてNK細胞の培養を行った結果からは、培養液と添加物によって増幅効率が変わることが分かった。しかし、EasySep Human NK Cell Enrichment Kitを使用して単離したNK細胞を用いた場合には、十分な増幅効率は得られなかった。 The results of culturing NK cells using RPMI medium, KBM medium, or NTI medium showed that the expansion efficiency varied depending on the culture medium and additives. However, when using NK cells isolated using the EasySep Human NK Cell Enrichment Kit, sufficient expansion efficiency was not obtained.
B).実施例2(ドナーMixB,シングルドナーA)
培養原材料となる細胞の条件を変えて、培養試験を行った。
B). Example 2 (Donor Mix B, Single Donor A)
Cultivation tests were conducted by changing the conditions of the cells used as the culture material.
・Day0
凍結保存PBMCを所定の方法で解凍し、以下の4群に調製した細胞懸濁液を12ウェルプレートに0.65mL播種し培養を開始した。
(1)PBMC(CD3−,CD34−)4x105cells/mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(2)PBMC(CD3−,CD34−)4x105cells/mL,RPMI培地※2,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)PBMC(CD3+,CD34+)4x105cells/mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(4)PBMC(CD3+,CD34+)4x105cells/mL,RPMI培地※2,Urelumab[1μg/mL],IL−21[1ng/mL]
・Day 0
Cryopreserved PBMCs were thawed by a prescribed method, and 0.65 mL of the cell suspension prepared for the following four groups was seeded onto a 12-well plate to initiate culture.
(1) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(2) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, RPMI medium *2 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(3) PBMC (CD3 + , CD34 + ) 4x10 5 cells/mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(4) PBMC (CD3 + , CD34 + ) 4x10 5 cells/mL, RPMI medium *2 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day6
各ウェルをよくピペッティングし細胞数をカウントした。それぞれの群を以下の条件の下、12ウェルプレートに1mL播種し継代培養した。
(1)−1 (1)細胞懸濁液4x105cells/1mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(2)−1 (2)細胞懸濁液4x105cells/1mL,RPMI培地※2,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)−1 (3)細胞懸濁液4x105cells/1mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(4)−1 (4)細胞懸濁液4x105cells/1mL,RPMI培地※2,Urelumab[1μg/mL],IL−21[1ng/mL]
Day 6
Each well was thoroughly pipetted and the number of cells was counted. Each group was seeded in 1 mL of a 12-well plate under the following conditions and subcultured.
(1)-1 (1) Cell suspension 4x10 5 cells/1 mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(2)-1 (2) Cell suspension 4x10 5 cells/1 mL, RPMI medium *2 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(3)-1 (3) Cell suspension 4x10 5 cells/1 mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(4)-1 (4) Cell suspension 4x10 5 cells/1 mL, RPMI medium *2 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day7
各ウェルをよくピペッティングし、それぞれの群を以下の条件の下、6ウェルプレートに2.0mL播種し継代培養した。
(1)−2 (1)−1細胞懸濁液1mL,1.0mL Urelumab[1μg/mL]添加済NTI培地※1
(2)−2 (2)−1細胞懸濁液1mL,1.0mL Urelumab[1μg/mL]添加済RPMI培地※2
(3)−2 (3)−1細胞懸濁液1mL,1.0mL Urelumab[1μg/mL]添加済NTI培地※1
(4)−2 (4)−1細胞懸濁液1mL,1.0mL Urelumab[1μg/mL]添加済RPMI培地※2
Day 7
Each well was thoroughly pipetted, and 2.0 mL of each group was inoculated into a 6-well plate under the following conditions and subcultured.
(1)-2 (1)-1 Cell suspension 1 mL, 1.0 mL Urelumab [1 μg/mL]-supplemented NTI medium *1
(2)-2 (2)-1 Cell suspension 1 mL, 1.0 mL Urelumab [1 μg/mL]-supplemented RPMI medium *2
(3)-2 (3)-1 Cell suspension 1 mL, 1.0 mL Urelumab [1 μg/mL]-supplemented NTI medium *1
(4)-2 (4)-1 Cell suspension 1 mL, 1.0 mL Urelumab [1 μg/mL]-supplemented RPMI medium *2
・Day10
各ウェルをよくピペッティングし細胞数をカウントした。それぞれの群を以下の条件の下、6ウェルプレートに3.0mL播種し継代培養した。
(1)−3 (1)−2細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(2)−3 (2)−2細胞懸濁液5x105cells/3mL,RPMI培地※2,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)−3 (3)−2細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(4)−3 (4)−2細胞懸濁液5x105cells/3mL,RPMI培地※2,Urelumab[1μg/mL],IL−21[1ng/mL]
・Day 10
Each well was thoroughly pipetted and the number of cells was counted. Each group was seeded in 3.0 mL of a 6-well plate under the following conditions and subcultured.
(1)-3 (1)-2 Cell suspension 5x10 5 cells/3mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(2)-3 (2)-2 Cell suspension 5x10 5 cells/3 mL, RPMI medium *2 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(3)-3 (3)-2 Cell suspension 5x10 5 cells/3mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(4)-3 (4)-2 Cell suspension 5x10 5 cells/3 mL, RPMI medium *2 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day12
(1)−3、(2)−3、(3)−3、(4)−3の各ウェルをよくピペッティングし細胞数をカウントした。
・Day 12
The cells in each of the wells (1)-3, (2)-3, (3)-3, and (4)-3 were thoroughly pipetted and counted.
・Day14
各ウェルをよくピペッティングし細胞数をカウントした。(1)、(3)、(4)の群をそれぞれ以下の条件の下、6ウェルプレートに3.0mL播種し継代培養した。
(1)−4 (1)−3細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)−4 (3)−3細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(4)−4 (4)−3細胞懸濁液5x105cells/3mL,RPMI培地※2,Urelumab[1μg/mL],IL−21[1ng/mL]
・Day 14
Each well was thoroughly pipetted and the number of cells was counted. Groups (1), (3), and (4) were seeded in 3.0 mL portions onto 6-well plates under the following conditions and subcultured.
(1)-4 (1)-3 Cell suspension 5x10 5 cells/3mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(3)-4 (3)-3 Cell suspension 5x10 5 cells/3mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(4)-4 (4)-3 Cell suspension 5x10 5 cells/3 mL, RPMI medium *2 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day17
各ウェルをよくピペッティングし細胞数をカウントした。(1)、(3)、(4)の群をそれぞれ以下の条件の下、6ウェルプレートに3.0mL播種し継代培養した。
(1)−5 (1)−4細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)−5 (3)−4細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(4)−5 (4)−4細胞懸濁液5x105cells/3mL,RPMI培地※2,Urelumab[1μg/mL],IL−21[1ng/mL]
・Day 17
Each well was thoroughly pipetted and the number of cells was counted. Groups (1), (3), and (4) were seeded in 3.0 mL portions onto 6-well plates under the following conditions and subcultured.
(1)-5 (1)-4 Cell suspension 5x10 5 cells/3mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(3)-5 (3)-4 Cell suspension 5x10 5 cells/3 mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(4)-5 (4)-4 Cell suspension 5x10 5 cells/3 mL, RPMI medium *2 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day19
(1)−5、(3)−5、(4)−5の各ウェルをよくピペッティングし細胞数をカウントした。
・Day 19
The cells in each of wells (1)-5, (3)-5, and (4)-5 were thoroughly pipetted and counted.
・Day21
(1)−5、(3)−5、(4)−5の各ウェルをよくピペッティングし細胞数をカウントした。(3)−5をそれぞれ以下の条件の下、6ウェルプレートに3.0mL播種し継代培養した。
(3)−5−1 (3)−5細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)−5−2 (3)−5細胞懸濁液1.5x106cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
・Day 21
The number of cells in each of the wells (1)-5, (3)-5, and (4)-5 was counted by pipetting thoroughly. 3.0 mL of each of the wells (3)-5 was seeded in a 6-well plate under the following conditions and subcultured.
(3)-5-1 (3)-5 Cell suspension 5x10 5 cells/3mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(3)-5-2 (3)-5 cell suspension 1.5x10 6 cells/3mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL]
・Day24
(3)−5−1、(3)−5−2の各ウェルをよくピペッティングし細胞数をカウントした。(3)−5−1、(3)−5−2をそれぞれ以下の条件の下、6ウェルプレートに3.0mL播種し継代培養した。
(3)−5−1 (3)−5−1細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)−5−2−a (3)−5−2細胞懸濁液5x105cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)−5−2−b (3)−5−2細胞懸濁液1.5x106cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
・Day 24
The number of cells in each well of (3)-5-1 and (3)-5-2 was counted by pipetting thoroughly. (3)-5-1 and (3)-5-2 were each seeded in 3.0 mL portions into a 6-well plate under the following conditions, and subcultured.
(3)-5-1 (3)-5-1 Cell suspension 5x10 5 cells/3 mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(3)-5-2-a (3)-5-2 cell suspension 5x10 5 cells/3mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(3)-5-2-b (3)-5-2 cell suspension 1.5x10 6 cells/3 mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day28
(3)−5−1、(3)−5−2−a、(3)−5−2−b、各ウェルをよくピペッティングし細胞数をカウントした。(3)−5−2−bを以下の条件の下、6ウェルプレートに3.0mL播種し継代培養した。
(3)−5−2−b’ (3)−5−2−b細胞懸濁液1.5x106cells/3mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
・Day 28
Each well of (3)-5-1, (3)-5-2-a, and (3)-5-2-b was thoroughly pipetted and the number of cells was counted. (3)-5-2-b was seeded in 3.0 mL in a 6-well plate under the following conditions and subcultured.
(3)-5-2-b' (3)-5-2-b cell suspension 1.5x10 6 cells/3 mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day31
(3)−5−2−b’のウェルをよくピペッティングし細胞数をカウントした。
・Day 31
(3)-5-2-b' well was thoroughly pipetted and the number of cells was counted.
図2に細胞の増殖倍率の変化を示す。NTI培地を用いる場合には、RPMI培地を用いる場合と比較して、細胞の増幅効率が良好となることが分かった。 Figure 2 shows the change in cell proliferation rate. It was found that the cell proliferation efficiency was better when NTI medium was used than when RPMI medium was used.
(1)、(3)、(4)について、各細胞表面マーカーについて実施したフローサイトメトリーの結果を図3に示す。得られた細胞集団には、特許文献1等に記載される高い傷害活性を示す細胞と同様の表現型を有する細胞が多く含まれていたが、一方で、CD3陽性細胞も増幅されてしまった。 The results of flow cytometry performed on each cell surface marker for (1), (3), and (4) are shown in Figure 3. The obtained cell population contained many cells with a phenotype similar to the cells exhibiting high cytotoxicity described in Patent Document 1, etc., but CD3-positive cells were also amplified.
C).実施例3(ドナーMixB)
抗4−1BB抗体とIL−21の濃度を変えて培養試験を行った。
C). Example 3 (Donor Mix B)
A culture test was carried out using different concentrations of anti-4-1BB antibody and IL-21.
・Day0
凍結保存PBMCを所定の方法で解凍し、以下の12群に調製した細胞懸濁液を12ウェルプレートに0.65mL播種し培養を開始した。
(1)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3
(2)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,IL−21[1ng/mL]
(3)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,IL−21[10ng/mL]
(4)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,IL−21[100ng/mL]
(5)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,Urelumab[1μg/mL]
(6)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,Urelumab[1μg/mL],IL−21[1ng/mL]
(7)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,Urelumab[1μg/mL],IL−21[10ng/mL]
(8)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,Urelumab[1μg/mL],IL−21[100ng/mL]
(9)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,Urelumab[10μg/mL],
(10)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,Urelumab[10μg/mL],IL−21[1ng/mL]
(11)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,Urelumab[10μg/mL],IL−21[10ng/mL]
(12)PBMC(CD3−,CD34−)4x105cells/mL,KBM502培地※3,Urelumab[10μg/mL],IL−21[100ng/mL]
・Day 0
Cryopreserved PBMCs were thawed by a prescribed method, and 0.65 mL of the cell suspension prepared for the following 12 groups was seeded onto a 12-well plate to initiate culture.
(1) PBMC ( CD3- , CD34- ) 4x105 cells/mL, KBM502 medium *3
(2) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , IL-21 [1 ng/mL]
(3) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , IL-21 [10 ng/mL]
(4) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , IL-21 [100 ng/mL]
(5) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , Urelumab [1 μg/mL]
(6) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(7) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [10 ng/mL]
(8) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [100 ng/mL]
(9) PBMC ( CD3- , CD34- ) 4x105 cells/mL, KBM502 medium *3 , Urelumab [10μg/mL],
(10) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , Urelumab [10 μg/mL], IL-21 [1 ng/mL]
(11) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , Urelumab [10 μg/mL], IL-21 [10 ng/mL]
(12) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM502 medium *3 , Urelumab [10 μg/mL], IL-21 [100 ng/mL]
また、以下の濃度に調製した細胞懸濁液をT75フラスコに13.5mL播種し培養を開始した。
(13)PBMC(CD3−,CD34−)5x105cells/mL,KBM502培地※3
Furthermore, 13.5 mL of the cell suspension prepared to the following concentration was seeded in a T75 flask to initiate culture.
(13) PBMC ( CD3- , CD34- ) 5x105 cells/mL, KBM502 medium *3
・Day6
(6)、(7)、(8)の各ウェルをよくピペッティングし細胞数をカウントした。以下の条件の下、12ウェルプレートに0.65mL播種し継代培養した。また、(6)、(7)、(8)の各ウェルには新しいKBM502培地※3を0.6mL追加した。
(6)−1 (6)細胞懸濁液0.25mL,0.4mLKBM502培地※3,Urelumab[1μg/mL],
(6)−2 (6)細胞懸濁液0.25mL,0.4mL KBM502培地※3,Urelumab[1μg/mL],IL−21[1ng/mL]
(7)−1 (7)細胞懸濁液0.25mL,0.4mL KBM502培地※3,Urelumab[1μg/mL],
(7)−2 (7)細胞懸濁液0.25mL,0.4mL KBM502培地※3,Urelumab[1μg/mL],IL−21[10ng/mL]
(8)−1 (8)細胞懸濁液0.25mL,0.4mL KBM502培地※3,Urelumab[1μg/mL],
(8)−2 (8)細胞懸濁液0.25mL,0.4mL KBM502培地※3,Urelumab[1μg/mL],IL−21[100ng/mL]
Day 6
The number of cells was counted by pipetting each of wells (6), (7), and (8). 0.65 mL of the cells were seeded in a 12-well plate under the following conditions and subcultured. In addition, 0.6 mL of fresh KBM502 medium *3 was added to each of wells (6), (7), and (8).
(6)-1 (6) Cell suspension 0.25 mL, 0.4 mL KBM502 medium *3 , Urelumab [1 μg / mL],
(6)-2 (6) Cell suspension 0.25 mL, 0.4 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(7)-1 (7) Cell suspension 0.25 mL, 0.4 mL KBM502 medium *3 , Urelumab [1 μg/mL],
(7)-2 (7) Cell suspension 0.25 mL, 0.4 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [10 ng/mL]
(8)-1 (8) Cell suspension 0.25 mL, 0.4 mL KBM502 medium *3 , Urelumab [1 μg/mL],
(8)-2 (8) Cell suspension 0.25 mL, 0.4 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [100 ng/mL]
・Day7
(1)から(12)までの各ウェルをよくピペッティングし細胞数をカウントした。さらに(10)、(11)、(12)、のそれぞれのウェルにおいて、以下の条件の下、12ウェルプレートに0.65mL播種し継代培養した。
(10)−1 (10)細胞懸濁液0.1mL,0.55mL KBM502培地※3,
(10)−2 (10)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL]
(10)−3 (10)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[1ng/mL]
(10)−4 (10)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[10ng/mL]
(10)−5 (10)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[100ng/mL]
Day 7
The number of cells was counted by pipetting each of wells (1) to (12). Furthermore, 0.65 mL of each of wells (10), (11), and (12) was seeded in a 12-well plate under the following conditions and subcultured.
(10)-1 (10) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 ,
(10)-2 (10) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg / mL]
(10)-3 (10) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(10)-4 (10) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [10 ng/mL]
(10)-5 (10) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [100 ng/mL]
(11)−1 (11)細胞懸濁液0.1mL,0.55mL KBM502培地※3
(11)−2 (11)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL]
(11)−3 (11)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[1ng/mL]
(11)−4 (11)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[10ng/mL]
(11)−5 (11)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[100ng/mL]
(11)-1 (11) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3
(11)-2 (11) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL]
(11)-3 (11) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(11)-4 (11) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [10 ng/mL]
(11)-5 (11) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [100 ng/mL]
(12)−1 (12)細胞懸濁液0.1mL,0.55mL KBM502培地※3
(12)−2 (12)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL]
(12)−3 (12)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[1ng/mL]
(12)−4 (12)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[10ng/mL]
(12)−5 (12)細胞懸濁液0.1mL,0.55mL KBM502培地※3,Urelumab[1μg/mL],IL−21[100ng/mL]
(12)-1 (12) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3
(12)-2 (12) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL]
(12)-3 (12) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(12)-4 (12) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [10 ng/mL]
(12)-5 (12) Cell suspension 0.1 mL, 0.55 mL KBM502 medium *3 , Urelumab [1 μg/mL], IL-21 [100 ng/mL]
・Day8
Day6で継代培養した(6)−1、(6)−2、(7)−1、(7)−2、(8)−1、(8)−2の各ウェルをよくピペッティングし細胞数をカウントした。以下の条件の下、6ウェルプレートに1.8mL播種し継代培養した。
(6)−1−1 (6)−1細胞懸濁液0.6mL,1.2mL KBM502培地※3
(6)−2−1 (6)−2細胞懸濁液0.6mL,1.2mL KBM502培地※3
(7)−1−1 (7)−1細胞懸濁液0.6mL,1.2mL KBM502培地※3
(7)−2−1 (7)−2細胞懸濁液0.6mL,1.2mL KBM502培地※3
(8)−1−1 (8)−1細胞懸濁液0.6mL,1.2mL KBM502培地※3
(8)−2−1 (8)−2細胞懸濁液0.6mL,1.2mL KBM502培地※3
・Day 8
The number of cells was counted by thoroughly pipetting each of the wells (6)-1, (6)-2, (7)-1, (7)-2, (8)-1, and (8)-2 that had been subcultured on Day 6. 1.8 mL of the cells were seeded in a 6-well plate and subcultured under the following conditions.
(6)-1-1 (6)-1 Cell suspension 0.6 mL, 1.2 mL KBM502 medium *3
(6)-2-1 (6)-2 Cell suspension 0.6 mL, 1.2 mL KBM502 medium *3
(7)-1-1 (7)-1 Cell suspension 0.6 mL, 1.2 mL KBM502 medium *3
(7)-2-1 (7)-2 Cell suspension 0.6 mL, 1.2 mL KBM502 medium *3
(8)-1-1 (8)-1 Cell suspension 0.6 mL, 1.2 mL KBM502 medium *3
(8)-2-1 (8)-2 Cell suspension 0.6 mL, 1.2 mL KBM502 medium *3
・Day10
Day8で継代培養した(6)−1−1、(6)−2−1、(7)−1−1、(7)−2−1、(8)−1−1、(8)−2−1の各ウェルをよくピペッティングし細胞数をカウントした。以下の条件の下、10cmディッシュに10mL播種し継代培養した。
(6)−1−2 (6)−1−1細胞懸濁液1.5mL,8.5mL KBM502培地※3
(6)−2−2 (6)−2−1細胞懸濁液1.5mL,8.5mL KBM502培地※3
(7)−1−2 (7)−1−1細胞懸濁液1.5mL,8.5mL KBM502培地※3
(7)−2−2 (7)−2−1細胞懸濁液1.5mL,8.5mL KBM502培地※3
(8)−1−2 (8)−1−1細胞懸濁液1.5mL,8.5mL KBM502培地※3
(8)−2−2 (8)−2−1細胞懸濁液1.5mL,8.5mL KBM502培地※3
・Day 10
The number of cells was counted by thorough pipetting in each well of (6)-1-1, (6)-2-1, (7)-1-1, (7)-2-1, (8)-1-1, and (8)-2-1 that had been subcultured on Day 8. 10 mL of the cells were seeded in a 10 cm dish and subcultured under the following conditions.
(6)-1-2 (6)-1-1 Cell suspension 1.5mL, 8.5mL KBM502 medium *3
(6)-2-2 (6)-2-1 Cell suspension 1.5mL, 8.5mL KBM502 medium *3
(7)-1-2 (7)-1-1 Cell suspension 1.5mL, 8.5mL KBM502 medium *3
(7)-2-2 (7)-2-1 Cell suspension 1.5mL, 8.5mL KBM502 medium *3
(8)-1-2 (8)-1-1 Cell suspension 1.5mL, 8.5mL KBM502 medium *3
(8)-2-2 (8)-2-1 Cell suspension 1.5mL, 8.5mL KBM502 medium *3
Day0で播種した(1)、(2)、(3)、(4)、(5)、(9)の各ウェルをよくピペッティングし細胞数をカウントした。
(1)のウェルには新しいKBM502培地※3を0.7mL追加した。
The cells seeded on Day 0 in each of wells (1), (2), (3), (4), (5), and (9) were thoroughly pipetted and the number of cells was counted.
0.7 mL of fresh KBM502 medium *3 was added to well (1).
Day7で播種した(10)−1、(10)−2、(10)−3、(10)−4、(10)−5、(11)−1、(11)−2、(11)−3、(11)−4、(11)−5、(12)−1、(12)−2、(12)−3、(12)−4、(12)−5、の各ウェルをよくピペッティングし細胞数をカウントした。以下の条件の下、6ウェルプレートに2.0mL播種し継代培養した。
(10)−1−1 (10)−1細胞懸濁液0.5mL,1.5mL KBM502培地※3
(11)−1−1 (11)−1細胞懸濁液0.5mL,1.5mL KBM502培地※3
(12)−1−1 (12)−1細胞懸濁液0.5mL,1.5mL KBM502培地※3
The number of cells was counted by pipetting thoroughly in each well of (10)-1, (10)-2, (10)-3, (10)-4, (10)-5, (11)-1, (11)-2, (11)-3, (11)-4, (11)-5, (12)-1, (12)-2, (12)-3, (12)-4, and (12)-5 seeded on Day 7. 2.0 mL was seeded in a 6-well plate and subcultured under the following conditions.
(10)-1-1 (10)-1 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
(11)-1-1 (11)-1 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
(12)-1-1 (12)-1 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
(10)−2−1 (10)−2細胞懸濁液0.5mL,1.5mL KBM502培地※3
(11)−2−1 (11)−2細胞懸濁液0.5mL,1.5mL KBM502培地※3
(12)−2−1 (12)−2細胞懸濁液0.5mL,1.5mL KBM502培地※3
(10)-2-1 (10)-2 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
(11)-2-1 (11)-2 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
(12)-2-1 (12)-2 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
(10)−3−1 (10)−3細胞懸濁液0.5mL,1.5mL KBM502培地※3
(11)−4−1 (11)−4細胞懸濁液0.5mL,1.5mL KBM502培地※3
(12)−5−1 (12)−5細胞懸濁液0.5mL,1.5mL KBM502培地※3
(10)-3-1 (10)-3 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
(11)-4-1 (11)-4 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
(12)-5-1 (12)-5 Cell suspension 0.5mL, 1.5mL KBM502 medium *3
・Day13
Day10で継代培養した(10)−1−1、(11)−1−1、(12)−1−1、(10)−2−1、(11)−2−1、(12)−2−1、(10)−3−1、(11)−4−1、(12)−5−1の各ウェルをよくピペッティングし細胞数をカウントした。以下の条件の下、6ウェルプレートに2.0mL播種し継代培養した。
(10)−1−2 (10)−1−1細胞懸濁液5x105cells/2mL,Urelumab[1μg/mL]添加済KBM502培地※3
(10)−2−2 (10)−2−1細胞懸濁液5x105cells/2mL,Urelumab[1μg/mL]添加済KBM502培地※3
(10)−3−2 (10)−3−1細胞懸濁液5x105cells/2mL,Urelumab[1μg/mL]添加済KBM502培地※3
(11)−2−2 (11)−2−1細胞懸濁液5x105cells/2mL,Urelumab[1μg/mL]添加済KBM502培地※3
・Day 13
The number of cells was counted by thoroughly pipetting each well of (10)-1-1, (11)-1-1, (12)-1-1, (10)-2-1, (11)-2-1, (12)-2-1, (10)-3-1, (11)-4-1, and (12)-5-1 that had been subcultured on Day 10. 2.0 mL of each well was seeded in a 6-well plate and subcultured under the following conditions.
(10)-1-2 (10)-1-1 Cell suspension 5x105 cells/2mL, KBM502 medium supplemented with Urelumab [1μg/mL] *3
(10)-2-2 (10)-2-1 Cell suspension 5x105 cells/2mL, KBM502 medium supplemented with Urelumab [1μg/mL] *3
(10)-3-2 (10)-3-1 Cell suspension 5x105 cells/2mL, KBM502 medium supplemented with Urelumab [1μg/mL] *3
(11)-2-2 (11)-2-1 Cell suspension 5x105 cells/2mL, KBM502 medium supplemented with Urelumab [1μg/mL] *3
(10)−1−3 (10)−1−1細胞懸濁液5x105cells/2mL,KBM502培地※3
(10)−2−3 (10)−2−1細胞懸濁液5x105cells/2mL,KBM502培地※3
(10)−3−3 (10)−3−1細胞懸濁液5x105cells/2mL,KBM502培地※3
(11)−2−3 (11)−2−1細胞懸濁液5x105cells/2mL,KBM502培地※3
(10)-1-3 (10)-1-1 Cell suspension 5x105 cells/2mL, KBM502 medium *3
(10)-2-3 (10)-2-1 Cell suspension 5x105 cells/2mL, KBM502 medium *3
(10)-3-3 (10)-3-1 Cell suspension 5x105 cells/2mL, KBM502 medium *3
(11)-2-3 (11)-2-1 Cell suspension 5x105 cells/2mL, KBM502 medium *3
・Day14
Day0で培養開始した(13)のT75フラスコをよくピペッティングし細胞数をカウントした。
・Day 14
On Day 0, the T75 flask in which the culture had been started (13) was thoroughly pipetted and the number of cells was counted.
Day10で継代培養した(6)−1−2、(6)−2−2、(7)−1−2、(7)−2−2、(8)−1−2、(8)−2−2の各10cmディッシュをよくピペッティングし細胞数をカウントした。以下の条件の下、6ウェルプレートに2.0mL播種し継代培養した。
(6)−a (6)−1−2細胞懸濁液5x105cells/2mL,KBM502培地※3
(6)−b (6)−1−2細胞懸濁液5x105cells/2mL,KBM502培地※3,Urelumab[1μg/mL]
(6)−c (6)−1−2細胞懸濁液5x105cells/2mL,KBM502培地※3,Urelumab[1μg/mL],IL−21[1ng/mL]
(6)−d (6)−1−2細胞懸濁液5x105cells/2mL,KBM501培地※4
The number of cells was counted by thoroughly pipetting each of the 10 cm dishes of (6)-1-2, (6)-2-2, (7)-1-2, (7)-2-2, (8)-1-2, and (8)-2-2 that had been subcultured on Day 10. 2.0 mL of each was seeded in a 6-well plate and subcultured under the following conditions.
(6)-a (6)-1-2 Cell suspension 5x105 cells/2mL, KBM502 medium *3
(6)-b (6)-1-2 Cell suspension 5x105 cells/2mL, KBM502 medium *3 , Urelumab [1μg/mL]
(6)-c (6)-1-2 cell suspension 5x105 cells/2mL, KBM502 medium *3 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(6)-d (6)-1-2 Cell suspension 5x105 cells/2mL, KBM501 medium *4
・Day15
Day13で継代培養した(10)−1−2、(10)−2−2、(10)−3−2、(11)−2−2、(10)−1−3、(10)−2−3、(10)−3−3、(11)−2−3の各ウェルをよくピペッティングし細胞数をカウントした。
・Day 15
On Day 13, each of the wells (10)-1-2, (10)-2-2, (10)-3-2, (11)-2-2, (10)-1-3, (10)-2-3, (10)-3-3, and (11)-2-3 that had been subcultured was thoroughly pipetted and the number of cells was counted.
・Day17
Day13で継代培養した(10)−1−2、(10)−2−2、(10)−3−2、(11)−2−2、(10)−1−3、(10)−2−3、(10)−3−3、(11)−2−3の各ウェルをよくピペッティングし細胞数をカウントした。以下の条件の下、6ウェルプレートに2.0mL播種し継代培養した。
(10)−1−3’ (10)−1−3細胞懸濁液1.0mL,1.0mL KBM502培地※3
(11)−2−2’ (11)−2−2細胞懸濁液1.0mL,1.0mL KBM502培地※3
・Day 17
The number of cells was counted by thorough pipetting in each well of (10)-1-2, (10)-2-2, (10)-3-2, (11)-2-2, (10)-1-3, (10)-2-3, (10)-3-3, and (11)-2-3 that had been subcultured on Day 13. 2.0 mL of the cells were seeded in a 6-well plate and subcultured under the following conditions.
(10)-1-3' (10)-1-3 Cell suspension 1.0 mL, 1.0 mL KBM502 medium *3
(11)-2-2' (11)-2-2 Cell suspension 1.0 mL, 1.0 mL KBM502 medium *3
Day14で継代培養した(6)−a、(6)−b、(6)−c、(6)−dの各ウェルをよくピペッティングし細胞数をカウントした。以下の条件の下、6ウェルプレートに2.0mL播種し継代培養した。
(6)−a’ (6)−a細胞懸濁液1.0mL,1.0mL KBM502培地※3
(6)−c’ (6)−c細胞懸濁液1.0mL,1.0mL KBM502培地※3
The number of cells was counted by thoroughly pipetting each of the wells (6)-a, (6)-b, (6)-c, and (6)-d that had been subcultured on Day 14. 2.0 mL of the cells were seeded in a 6-well plate and subcultured under the following conditions.
(6)-a' (6)-a Cell suspension 1.0 mL, 1.0 mL KBM502 medium *3
(6)-c' (6)-c Cell suspension 1.0 mL, 1.0 mL KBM502 medium *3
・Day20
Day13で継代培養した(10)−1−2、(10)−2−2、(10)−3−2、(10)−2−3、(10)−3−3、(11)−2−3の各ウェルをよくピペッティングし細胞数をカウントした。
Day14で継代培養した(6)−b、(6)−dの各ウェルをよくピペッティングし細胞数をカウントした。
・Day 20
On Day 13, each of the wells (10)-1-2, (10)-2-2, (10)-3-2, (10)-2-3, (10)-3-3, and (11)-2-3 that had been subcultured was thoroughly pipetted and the number of cells was counted.
On Day 14, the cells in each of the wells (6)-b and (6)-d that had been subcultured were thoroughly pipetted and the number of cells was counted.
・Day21
Day17で継代培養した(10)−1−3’、(11)−2−2’、(6)−a’、(6)−c’の各ウェルをよくピペッティングし細胞数をカウントした。
・Day 21
On Day 17, the cells in each of the wells (10)-1-3', (11)-2-2', (6)-a', and (6)-c' that had been subcultured were thoroughly pipetted and the number of cells was counted.
結果を表2(細胞の全体倍率の変化)、図4(細胞数の変化)および図5(増幅倍率)に示す。表2において、Day0を1とした場合のDay10以降の最後のカウント時の各細胞数のFold Changeの値により、細胞増幅結果を以下のようにA~Dと評価した。なお、評価がC以上の場合に、効率的な細胞の増幅が得られ、目的が達成されたと判断することができる。
A:Fold Changeが100以上
B:Fold Changeが10以上
C:Fold Changeが2以上
D:Fold Changeが2未満
The results are shown in Table 2 (change in total cell fold), Fig. 4 (change in cell number) and Fig. 5 (amplification fold). In Table 2, the cell amplification results were evaluated as A to D as follows, based on the fold change value of each cell number at the final count on Day 10 or later, with Day 0 being set as 1. When the evaluation is C or higher, it can be determined that efficient cell amplification has been obtained and the purpose has been achieved.
A: Fold change is 100 or more B: Fold change is 10 or more C: Fold change is 2 or more D: Fold change is less than 2
(2)~(4)の結果より、IL−21だけを添加して培養した場合には、細胞の増幅効率が良くなかった。(5)および(9)の結果より、IL−21を添加せずに培養した場合には、初動増殖が著しく悪かった。(6)~(8)および(10)~(12)の結果より、IL−21およびUrelumabを添加して培養した場合には、初動増殖が良好であった。 The results of (2) to (4) show that when cells were cultured with the addition of only IL-21, the cell expansion efficiency was poor. The results of (5) and (9) show that when cells were cultured without the addition of IL-21, the initial proliferation was significantly poor. The results of (6) to (8) and (10) to (12) show that when cells were cultured with the addition of IL-21 and Urelumab, the initial proliferation was good.
(6)−a、(6)−c、(10)−1−3または(11)−2−2の細胞集団について、各細胞表面マーカーについて実施したフローサイトメトリーの結果を図6に示す。 The results of flow cytometry performed on each cell surface marker for the cell populations (6)-a, (6)-c, (10)-1-3, or (11)-2-2 are shown in Figure 6.
D).実施例4(ドナーMixB)
得られた細胞集団の細胞傷害活性を確認した。
D). Example 4 (Donor Mix B)
The cytotoxic activity of the resulting cell population was confirmed.
・Day0
凍結保存PBMC(CD3−,CD34−)を所定の方法で解凍し、IL−18[100ng/mL]、Urelumab[1μg/mL]、IL−21[1ng/mL]を添加したNTI培地※1にて4x105cells/mLに調製した細胞懸濁液を6ウェルプレートに1.5mL播種し培養を開始した。また、Urelumab[1μg/mL]、IL−21[1ng/mL]を添加したNTI培地※1にて4x105cells/mLに調製した細胞懸濁液をT75フラスコ2個にそれぞれ12.5mL播種し培養を開始した。
・Day 0
Cryopreserved PBMC ( CD3- , CD34- ) were thawed by a prescribed method, and the cell suspension was adjusted to 4x105 cells/mL in NTI medium *1 supplemented with IL-18 [100ng/mL], Urelumab [1μg/mL], and IL-21 [1ng/mL], and 1.5mL of the cell suspension was seeded in a 6-well plate to start culture. In addition, 12.5mL of the cell suspension was seeded in two T75 flasks to start culture, and 4x105 cells/mL in NTI medium *1 supplemented with Urelumab [1μg/mL] and IL-21 [1ng/mL], and 12.5mL of the cell suspension was seeded in two T75 flasks to start culture.
・Day7
6ウェルプレートをよくピペッティングし、細胞数をカウントした。新しいNTI培地※1で2x105cells/mLに調製した細胞懸濁液にIL−18[100ng/mL]、Urelumab[1μg/mL]、IL−21[1ng/mL]を添加、T75フラスコAに14.4mL播種し継代培養した。
Day 7
The 6-well plate was pipetted thoroughly and the number of cells was counted. IL-18 [100 ng/mL], Urelumab [1 μg/mL], and IL- 21 [1 ng/mL] were added to the cell suspension prepared at 2 x 10 5 cells/mL in fresh NTI medium*1, and 14.4 mL of the mixture was seeded in a T75 flask A and subcultured.
T75フラスコ2個をよくピペッティングし、細胞をカウントした。新しいNTI培地※1で2x105cells/mLに調製した細胞懸濁液にUrelumab[1μg/mL]、IL−21[1ng/mL]を添加、T225フラスコ2個((1)、(2))に45mLずつ、T225フラスコ2個((3)、(4))に64mLずつ播種し継代培養した。 The two T75 flasks were pipetted thoroughly and the cells were counted. A cell suspension was prepared at 2 x 105 cells/mL in fresh NTI medium *1, to which Urelumab [1 μg/mL] and IL-21 [1 ng/mL] were added. The cells were then seeded in two T225 flasks ((1) and (2)) at 45 mL each and in two T225 flasks ((3) and (4)) at 64 mL each, and subcultured.
・Day10
T75フラスコAをよくピペッティングし、細胞をカウントした。新しいNTI培地※1で2x105cells/mLに調製した細胞懸濁液をT75フラスコBに15mL播種し継代培養した。回収後のT75フラスコAにはNTI培地※115mLを入れ継代培養した。
(1)、(2)のT225フラスコをよくピペッティングし、細胞をカウントした。以下の2群に調製し、T225フラスコ2個にそれぞれ播種し継代培養した。
(1)−1 (1)細胞懸濁液9x106cellsを分取、500g,5分遠心後新しいNTI培地※145mL(2x105cells/mL)で懸濁
(1)−2 (1)細胞懸濁液9x106cells/45mL,NTI培地※1
・Day 10
The T75 flask A was pipetted thoroughly and the cells were counted. 15 mL of the cell suspension adjusted to 2 x 10 5 cells/mL with fresh NTI medium *1 was seeded into T75 flask B and subcultured. 15 mL of NTI medium *1 was added to the recovered T75 flask A and subcultured.
The T225 flasks (1) and (2) were thoroughly pipetted and the cells were counted. The following two groups were prepared, seeded in two T225 flasks each, and subcultured.
(1)-1 (1) Cell suspension 9x106 cells were taken, centrifuged at 500g for 5 minutes, and then suspended in 45mL ( 2x105 cells/mL) of fresh NTI medium *1. (1)-2 (1) Cell suspension 9x106 cells/45mL, NTI medium *1
(3)、(4)のT225フラスコをよくピペッティングし、細胞数をカウントし回収した。
(1)、(2)、(3)、(4)それぞれ回収後のT225フラスコには50mLのNTI培地※115mLを入れ継代培養した。
The T225 flasks from (3) and (4) were thoroughly pipetted, and the cells were counted and recovered.
After recovery from (1), (2), (3), and (4), 15 mL of 50 mL of NTI medium *1 was added to each T225 flask and subcultured.
・Day14
(1)−1、(1)−2のT225フラスコをよくピペッティングし、細胞数をカウントした。以下の2群に調製し、T225フラスコ2個にそれぞれ播種し継代培養した。
(1)−1−1 (1)−1細胞懸濁液2.5x106cellsを分取、500g,5分遠心後新しいNTI培地※150mLで懸濁
(1)−2−1 (1)−1細胞懸濁液2.5x106cells/50mL,NTI培地※1
・Day 14
The T225 flasks (1)-1 and (1)-2 were thoroughly pipetted and the number of cells was counted. The following two groups were prepared, and each was seeded in two T225 flasks and subcultured.
(1)-1-1 (1)-1 Cell suspension 2.5x106 cells were taken, centrifuged at 500g for 5 minutes, and then suspended in 50mL of fresh NTI medium *1. (1)-2-1 (1)-1 Cell suspension 2.5x106 cells/50mL, NTI medium *1
A、BのT75フラスコをよくピペッティングし、細胞数をカウントした。以下の2群に調製し、6ウェルプレートにそれぞれ播種し継代培養した。
A−1 A細胞懸濁液1.0x106cells/2mL,NTI培地※1
B−1 A細胞懸濁液1.0x106cellsを分取、500g,5分遠心後新しいNTI培地※12mLで懸濁
The T75 flasks A and B were thoroughly pipetted and the number of cells was counted. The following two groups were prepared, seeded on 6-well plates, and subcultured.
A-1 A cell suspension 1.0x106 cells/2mL, NTI medium *1
B-1: Take 1.0x106 cells from the cell suspension A, centrifuge at 500g for 5 minutes, and then suspend in 2mL of fresh NTI medium *1.
・Day15
(1)、(2)、(3)、(4)のT225フラスコをよくピペッティングし、1mMEDTA/PBSを使用して細胞を回収し細胞数をカウントした。
新しいNTI培地※1で1.18x106cells/mLに調製した細胞懸濁液をT75フラスコ((5)、(6))2個に50mLずつ播種し継代培養した。
・Day 15
The T225 flasks from (1), (2), (3), and (4) were thoroughly pipetted, and the cells were recovered using 1 mM EDTA/PBS and the cell numbers were counted.
A cell suspension adjusted to 1.18 x 10 6 cells/mL in fresh NTI medium *1 was seeded at 50 mL each into two T75 flasks ((5) and (6)) and subcultured.
・Day16
(1)−1−1、(1)−2−1のT225フラスコ各1個をよくピペッティングし、1mMEDTA/PBSを使用して細胞を回収し細胞数をカウントした。
・Day 16
One T225 flask each of (1)-1-1 and (1)-2-1 was thoroughly pipetted, and the cells were recovered using 1 mM EDTA/PBS and the number of cells was counted.
・Day17
(1)−1−1、(1)−2−1のT225フラスコ各1個をよくピペッティングし、1mMEDTA/PBSを使用して細胞を回収し細胞数をカウントした。
A−1、B−1のウェルをよくピペッティングし、細胞数をカウントした。
・Day 17
One T225 flask each of (1)-1-1 and (1)-2-1 was thoroughly pipetted, and the cells were recovered using 1 mM EDTA/PBS and the number of cells was counted.
Wells A-1 and B-1 were thoroughly pipetted, and the number of cells was counted.
[腫瘍細胞に対する傷害活性測定]
細胞傷害活性の測定には、Day17に回収した細胞とK562細胞を反応させた群、陰性コントロールとしてK562細胞のみの群、陽性コントロールとしてK562細胞を10%ホルマリンで処理した群を用意した。
[Measurement of cytotoxicity against tumor cells]
For the measurement of cytotoxic activity, a group in which the cells collected on Day 17 were reacted with K562 cells, a group containing only K562 cells as a negative control, and a group in which K562 cells were treated with 10% formalin as a positive control were prepared.
《Day17回収細胞》
Day17回収細胞を、RPMI1640培地(10%FBSおよび1%ペニシリン−ストレプトマイシン混合溶液を添加)にて1x106cells/mlの濃度に調製した。
<<Day 17 Recovered Cells>>
The cells collected on Day 17 were adjusted to a concentration of 1 x 10 6 cells/ml in RPMI 1640 medium (containing 10% FBS and 1% penicillin-streptomycin mixed solution).
《K562》
K562細胞をRPMI1640培地(無血清)にて懸濁し、PKH26 Red Fluorescent Cell Linker Kitを用いて染色したのち、RPMI1640培地(10%FBSおよび1%ペニシリン−ストレプトマイシン混合溶液を添加)にて2x106cells/mLとなるように調製した。
《K562》
K562 cells were suspended in RPMI1640 medium (serum-free) and stained using PKH26 Red Fluorescent Cell Linker Kit, and then adjusted to 2 x 10 6 cells/mL in RPMI1640 medium (containing 10% FBS and 1% penicillin-streptomycin mixed solution).
Day17回収細胞とK562細胞は、細胞比で1:1および2:1となるように96ウェルプレート(IWAKI,4870−800SP)に添加、混合し、37℃、5% CO2下で2時間反応させた。反応後、500g,5分遠心し、上清を除去後、PBSで希釈した7−AAD溶液を添加、懸濁し、室温で20分間インキュベートした。フローサイトメーターを用いて測定を行い、FlowJoソフトウェアで解析し、細胞傷害活性率(%Lysis)を算出した。 Day 17 collected cells and K562 cells were added to a 96-well plate (IWAKI, 4870-800SP) at a cell ratio of 1:1 and 2:1, mixed, and reacted for 2 hours at 37°C under 5% CO2. After the reaction, the cells were centrifuged at 500g for 5 minutes, the supernatant was removed, and then 7-AAD solution diluted with PBS was added, suspended, and incubated at room temperature for 20 minutes. Measurement was performed using a flow cytometer, and analysis was performed using FlowJo software to calculate the cytotoxic activity rate (% Lysis).
細胞傷害活性率(%Lysis)=(K562細胞死細胞率−陰性コントロール死細胞率)/(陽性コントロール死細胞率−陰性コントロール死細胞率)×100 Cytotoxic activity rate (% Lysis) = (K562 cell death rate - negative control cell death rate) / (positive control cell death rate - negative control cell death rate) x 100
上記式で求められる細胞傷害活性率は、以下のようにA~Dで評価される。評価がC以上である場合に、高い細胞傷害活性が得られ、目的が達成されたと判断される。
A:70%以上
B:60%以上
C:50%以上
D:50%未満
The cytotoxic activity rate calculated by the above formula is rated as follows, from A to D. When the rating is C or higher, it is determined that high cytotoxic activity is obtained and the objective is achieved.
A: 70% or more B: 60% or more C: 50% or more D: Less than 50%
結果を図7に示す。E(エフェクター細胞(回収した細胞)):T(標的細胞(K562細胞))=1:1または2:1のいずれの場合においても、回収した細胞集団による細胞傷害活性率の評価はAであり、腫瘍細胞に対する特に高い傷害活性が示された。 The results are shown in Figure 7. In both cases where the ratio of E (effector cells (recovered cells)):T (target cells (K562 cells)) was 1:1 or 2:1, the cytotoxic activity rate of the recovered cell population was evaluated as A, indicating particularly high cytotoxic activity against tumor cells.
E).実施例5(ドナーMixB)
得られた細胞集団の抗腫瘍効果を確認した。
E). Example 5 (Donor Mix B)
The antitumor effect of the obtained cell population was confirmed.
・Day0
凍結保存PBMC(CD3−,CD34−)を所定の方法で解凍し、終濃度Urelumab[1μg/mL]、IL−21[1ng/mL]を添加したNTI培地※1にて4x105cells/mLに調製した細胞懸濁液をT75フラスコ2個に12.5mLずつ播種し培養を開始した。
・Day 0
Cryopreserved PBMCs (CD3 - , CD34 - ) were thawed using a prescribed method, and a cell suspension adjusted to 4 x 10 5 cells/mL in NTI medium *1 supplemented with final concentrations of Urelumab [1 μg/mL] and IL-21 [1 ng/mL] was seeded at 12.5 mL per flask into two T75 flasks to initiate culture.
・Day6
T75フラスコ2個から細胞懸濁液25mLを回収し、細胞をカウントした。新しいNTI培地※1を12.5mL追加、さらにUrelumab[1μg/mL]、IL−21[1ng/mL]を添加し、T75フラスコ3個に12.5mLずつ継代培養した。
Day 6
25 mL of cell suspension was collected from two T75 flasks and the cells were counted. 12.5 mL of fresh NTI medium *1 was added, and Urelumab [1 μg/mL] and IL-21 [1 ng/mL] were further added, and 12.5 mL each was subcultured in three T75 flasks.
・Day8
T75フラスコをよくピペッティングし、細胞数をカウントした。各々のフラスコをT225フラスコに拡大し、新しいNTI培地※1を37.5mL追加、さらにUrelumab[1μg/mL]、IL−21[1ng/mL]を添加して継代培養した。
・Day 8
The T75 flask was pipetted thoroughly and the number of cells was counted. Each flask was expanded to a T225 flask, and 37.5 mL of fresh NTI medium *1 was added, followed by the addition of Urelumab [1 μg/mL] and IL-21 [1 ng/mL] for subculture.
・Day10
T225フラスコをよくピペッティングし、細胞懸濁液を回収した。500g,5minで遠心し、ペレットを60mLの培養上清でサスペンドした。新しいNTI培地※1を240mL追加し、T225フラスコ4個に75mLずつ継代培養した。
・Day 10
The T225 flask was pipetted thoroughly to recover the cell suspension. It was centrifuged at 500g for 5 min, and the pellet was suspended in 60 mL of culture supernatant. 240 mL of fresh NTI medium *1 was added, and 75 mL each was subcultured in four T225 flasks.
・Day13
T225フラスコ1個をよくピペッティングし、細胞懸濁液を回収した。500g,5minで遠心し、細胞数をカウントした。
新しいNTI培地※1で9x105cells/mLに調製しT225フラスコ4個に50mLずつ継代培養した。
・Day 13
One T225 flask was thoroughly pipetted to recover the cell suspension, which was then centrifuged at 500 g for 5 minutes, and the number of cells was counted.
The cells were adjusted to 9 x 10 5 cells/mL in fresh NTI medium *1 and subcultured in 50 mL each in four T225 flasks.
・Day14
T225フラスコ1個をよくピペッティングし、1mM EDTA/PBSを使用して細胞を回収し細胞数をカウントした。
・Day 14
One T225 flask was thoroughly pipetted, and the cells were recovered using 1 mM EDTA/PBS, and the cell number was counted.
・Day15
T225フラスコ1個をよくピペッティングし、1mM EDTA/PBSを使用して細胞を回収し細胞数をカウントした。
・Day 15
One T225 flask was thoroughly pipetted, and the cells were recovered using 1 mM EDTA/PBS, and the cell number was counted.
・Day16
T225フラスコ1個をよくピペッティングし、1mM EDTA/PBSを使用して細胞を回収し細胞数をカウントした。
・Day 16
One T225 flask was thoroughly pipetted, and the cells were recovered using 1 mM EDTA/PBS, and the cell number was counted.
・Day17
T225フラスコ1個をよくピペッティングし、1mM EDTA/PBSを使用して細胞を回収し細胞数をカウントした。
・Day 17
One T225 flask was thoroughly pipetted, and the cells were recovered using 1 mM EDTA/PBS, and the cell number was counted.
[動物実験]
使用動物:
雌性NOD.Cg−PrkdcscidIl2rgtm1Sug/ShiJic(NOG)マウス(6週齢、日本クレア/実験動物中央研究所)を購入し、最大5日間の馴化飼育の後、6−7週齢で試験に用いた。動物は1ケージ当たり3−4頭で飼育し、温度:21~25℃、湿度:40~60%(エアコンによる空調制御)、照明時間:12時間(7:00~19:00)の環境に調節された動物飼育室で飼育し、固形飼料CRF−1(γ線滅菌済み、オリエンタル酵母株式会社)を自由摂取させた。飲水はオートクレーブ済み水道水を自由に摂取させた。
[Animal experiments]
Animals used:
Female NOD.Cg-Prkdc scid Il2rg tm1Sug /ShiJic (NOG) mice (6 weeks old, Japan CLEA/Central Institute for Experimental Animals) were purchased and used for the study at 6-7 weeks of age after acclimation for up to 5 days. The animals were housed in cages of 3-4 animals in an animal breeding room with an regulated environment of temperature: 21-25°C, humidity: 40-60% (air conditioning control), and lighting time: 12 hours (7:00-19:00), and were allowed to freely consume solid feed CRF-1 (γ-ray sterilized, Oriental Yeast Co., Ltd.). Autoclaved tap water was allowed to be consumed freely.
群構成:
下表の通りに、Control(無治療)群、および被験物質投与群の2群にて試験を行った。
Group composition:
As shown in the table below, the test was conducted on two groups: a control (untreated) group and a test substance administration group.
担がんマウスの作成:
《SK−OV−3/CMV−Luc細胞のsphere作成》
SK−OV−3/CMV−Luc細胞をRPMI1640培地(10%FBSおよび1%ペニシリン−ストレプトマイシン混合溶液を添加)にて1ウェルあたり1x106cells/3mlに調製し、EZSPHERE(登録商標)6ウェルプレート(IWAKI,4810−900SP)に播種した。72時間培養後、作成したsphere(2700spheres/well)を回収し100g,1分遠心し上清を除去し、PBSでやさしく懸濁し2700 spheres/3mLに調製した。
Creation of tumor-bearing mice:
<<Sphere creation of SK-OV-3/CMV-Luc cells>>
SK-OV-3/CMV-Luc cells were prepared in RPMI1640 medium (containing 10% FBS and 1% penicillin-streptomycin mixed solution) at 1x106 cells/3mL per well and seeded on an EZSPHERE (registered trademark) 6-well plate (IWAKI, 4810-900SP). After 72 hours of culture, the prepared spheres (2700 spheres/well) were collected and centrifuged at 100g for 1 minute to remove the supernatant, and gently suspended in PBS to prepare 2700 spheres/3mL.
SK−OV−3/CMV−Luc(ルシフェラーゼ発現ヒト卵巣がん細胞株)、450spheres/0.5mL/headをマウス腹腔内に27G注射針を使用し移植、腹膜播種モデルを作成した。 SK-OV-3/CMV-Luc (a luciferase-expressing human ovarian cancer cell line), 450 spheres/0.5 mL/head, was implanted into the abdominal cavity of mice using a 27G needle to create a peritoneal dissemination model.
被験物質:
回収細胞:自施設にて製造
ヒト・インターロイキン−2製剤(IL−2):イムネース(一般名:テセロイキン(遺伝子組換え))、塩野義製薬
Test substance:
Recovered cells: Produced in-house Human interleukin-2 preparation (IL-2): Imunace (generic name: Teceleukin (genetic recombinant)), Shionogi Pharmaceuticals
被験物質の投与:
SK−OV−3/CMV−Lucの移植日をDay0とし、Day3(1回目)、Day4(2回目)、Day5(3回目)、Day6(4回目)、Day7(5回目)に回収細胞を腹腔内投与した。
Test substance administration:
The day of SK-OV-3/CMV-Luc transplantation was designated as Day 0, and the recovered cells were intraperitoneally administered on Day 3 (1st), Day 4 (2nd), Day 5 (3rd), Day 6 (4th), and Day 7 (5th).
経過観察:
in vivoイメージングシステム(IVIS)による腹腔内腫瘍の蛍光観察
IVISによる観察ではLuciferin 3mg/head in PBS 200μLをマウス背部の皮下に投与し、腫瘍の発光を確認する。観察はイソフルラン麻酔下(動物用吸入麻酔器を使用、キャリアーガスに空気を用いイソフルラン濃度4%で導入、2%にて維持)で実施する。その後は1−2回/週、経時的に観察を行った。
Observation:
Fluorescence observation of intraperitoneal tumors using an in vivo imaging system (IVIS) For observation using IVIS, Luciferin 3 mg/head in 200 μL of PBS was administered subcutaneously to the back of the mouse, and the tumor was confirmed to emit light. Observation was performed under isoflurane anesthesia (using an animal inhalation anesthesia machine, with air as the carrier gas and isoflurane concentration 4% induced and maintained at 2%). After that, observation was performed 1-2 times/week over time.
体重測定および外観観察
SK−OV−3/CMV−Lucの移植後は、1−2回/週、体重測定と外観観察を行った。
Body Weight Measurement and Observation of Appearance After transplantation of SK-OV-3/CMV-Luc, body weight measurement and observation of appearance were performed 1-2 times a week.
人道的エンドポイント:
摂餌、摂水困難
Humane Endpoints:
Difficulty eating and drinking
結果を図8(SK−OV−3/CMV−Luc細胞数の変化)および図9(Day69のIVIS結果)に示す。回収した細胞集団を投与した群においては、抗腫瘍効果が認められた。 The results are shown in Figure 8 (changes in SK-OV-3/CMV-Luc cell count) and Figure 9 (IVIS results on Day 69). An antitumor effect was observed in the group administered the collected cell population.
F).実施例6(ドナーMixC)
・Day0
凍結保存PBMC(CD3−,CD34−)を所定の方法で解凍し、以下の条件の下培養を開始した。
(1)1.3x105cells/0.65mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]24ウェルプレート
(2)1.6x105cells/0.65mL,KBM501培地※4,24ウェルプレート
(3)1.3x105cells/0.65mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]G−Rex 24ウェルプレート
(4) 1.6x105cells/0.65mL,KBM501培地※4,G−Rex 24ウェルプレート
A 7.5x106cells/15mL,KBM501培地※4,T75フラスコ
B 7.5x106cells/15mL,KBM501培地※4,T75フラスコ
F). Example 6 (Donor Mix C)
・Day 0
Cryopreserved PBMC (CD3 − , CD34 − ) were thawed by a predetermined method, and culture was initiated under the following conditions.
(1) 1.3x105 cells/0.65mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL], 24-well plate (2) 1.6x105 cells/0.65mL, KBM501 medium *4 , 24-well plate (3) 1.3x105 cells/0.65mL, NTI medium *1 , Urelumab [1μg/mL], IL-21 [1ng/mL], G-Rex 24-well plate (4) 1.6x105 cells/0.65mL, KBM501 medium *4 , G-Rex 24-well plate A 7.5x106 cells/15mL, KBM501 medium *4 , T75 flask B 7.5x10 6 cells/15mL, KBM501 medium *4 , T75 flask
・Day7
(1)のウェルをよくピペッティングし、細胞数をカウントした。以下の条件の下、継代培養した。
(1)−1 (1)細胞懸濁液0.65mL,NTI培地※11.5mL,Urelumab[1μg/mL],IL−21[1ng/mL]
Day 7
The wells in (1) were thoroughly pipetted and the number of cells was counted. The cells were subcultured under the following conditions.
(1)-1 (1) Cell suspension 0.65 mL, NTI medium *1 1.5 mL, Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day10
T75フラスコAに新しいNTI培地※135mLを加え培養した。
T75フラスコBにUrelumab[1μg/mL]、IL−21[1ng/mL]を添加した新しいNTI培地※135mLを加え培養した。
(2)のウェルをよくピペッティングし、細胞数をカウントした。新しいKBM501培地※4を1mL加えた。
・Day 10
35 mL of fresh NTI medium *1 was added to T75 flask A and cultured.
35 mL of fresh NTI medium*1 supplemented with Urelumab [1 μg/mL] and IL-21 [1 ng/mL] was added to T75 flask B and cultured.
(2) Well was pipetted thoroughly and the number of cells was counted. 1 mL of fresh KBM501 medium *4 was added.
・Day11
(1)−1のウェルをよくピペッティングし、細胞数をカウントした。新しいNTI培地※1で6mLにメスアップ、Urelumab[1μg/mL]、IL−21[1ng/mL]を添加し、以下の3群に継代培養した。
(1)−2 (1)−1細胞懸濁液2mL
(1)−3 (1)−1細胞懸濁液2mL
(5)G−Rex 24ウェルプレート (1)−1細胞懸濁液2mL
また(1)−1の空のウェルには新しいNTI培地※12mLを加え継代培養した。
・Day 11
(1)-1 well was pipetted thoroughly and the number of cells was counted. Fresh NTI medium *1 was added to 6 mL, and Urelumab [1 μg/mL] and IL-21 [1 ng/mL] were added, and the cells were subcultured into the following three groups.
(1)-2 (1)-1 Cell suspension 2 mL
(1)-3 (1)-1 Cell suspension 2 mL
(5) G-Rex 24-well plate (1)-1 Cell suspension 2 mL
Furthermore, 2 mL of fresh NTI medium *1 was added to the empty well of (1)-1 and subcultured.
・Day13
T75フラスコAに新しいNTI培地※135mLを加え継代培養した。
(1)−2のウェルをよくピペッティングし、細胞数をカウントした。
(5)G−Rex 24ウェルプレートをよくピペッティングし、細胞数をカウントした。
・Day 13
35 mL of fresh NTI medium *1 was added to T75 flask A and subcultured.
The wells in (1)-2 were thoroughly pipetted, and the number of cells was counted.
(5) The G-Rex 24-well plate was thoroughly pipetted, and the number of cells was counted.
・Day14
(1)−2のウェルをよくピペッティングし、細胞数をカウントした。新しいNTI培地※1で6mLにメスアップ、2mLずつ6ウェルプレートに継代培養した。
(1)−2−1 (1)−2細胞懸濁液2mL
(1)−2−2 (1)−2細胞懸濁液2mL
(1)−2−3 (1)−2細胞懸濁液2mL
(1)−3のウェルをよくピペッティングし、細胞数をカウントした。以下の条件の下、6ウェルプレートに継代培養した。
(1)−3−1 (1)−3細棒懸濁液を500g,5分遠心後新しいUrelumab[1μg/mL],IL−21[1ng/mL]を添加したNTI培地※13mLでよく懸濁し播種
・Day 14
(1)-2 wells were thoroughly pipetted and the number of cells was counted. The cells were then diluted to 6 mL with fresh NTI medium *1 and subcultured in 2 mL portions onto 6-well plates.
(1)-2-1 (1)-2 Cell suspension 2 mL
(1)-2-2 (1)-2 Cell suspension 2 mL
(1)-2-3 (1)-2 Cell suspension 2 mL
The wells in (1)-3 were thoroughly pipetted and the number of cells was counted. The cells were subcultured in a 6-well plate under the following conditions.
(1)-3-1 (1)-3 The suspension of the thin rod was centrifuged at 500 g for 5 minutes, and then thoroughly suspended in 3 mL of NTI medium*1 containing fresh Urelumab [1 μg/mL] and IL-21 [1 ng/mL] and then inoculated.
(5)G−Rex 24ウェルをよくピペッティングし、細胞数をカウントし新しいNTI培地※1を4mL加え継代培養した。 (5) The G-Rex 24-well plate was thoroughly pipetted, the number of cells was counted, and 4 mL of fresh NTI medium *1 was added and the cells were subcultured.
T75フラスコAとBをよくピペッティングし、1mMEDTA/PBSを使用して細胞を回収し細胞数をカウントした。 T75 flasks A and B were thoroughly pipetted, and the cells were harvested using 1 mM EDTA/PBS and counted.
・Day17
(1)−2−1のウェルをよくピペッティングし、細胞数をカウントした。
(1)−3−1のウェルをよくピペッティングし、細胞数をカウントした。
(5)G−Rex 4ウェルから4.7mLの培地を静かに抜き取り、新しいNTI培地※1を4mL加えた。よくピペッティングし、細胞数をカウントした。さらに新しいNTI培地※1を1mL加えた。
・Day 17
The wells in (1)-2-1 were thoroughly pipetted, and the number of cells was counted.
The wells in (1)-3-1 were thoroughly pipetted, and the number of cells was counted.
(5) 4.7 mL of medium was gently removed from the G-Rex 4 wells, and 4 mL of fresh NTI medium *1 was added. The cells were thoroughly pipetted and counted. An additional 1 mL of fresh NTI medium *1 was added.
・Day20
(1)−2−1のウェルをよくピペッティングし、細胞数をカウントした。
(1)−3−1のウェルをよくピペッティングし、細胞数をカウントした。
・Day 20
The wells in (1)-2-1 were thoroughly pipetted, and the number of cells was counted.
The wells in (1)-3-1 were thoroughly pipetted, and the number of cells was counted.
・Day21
(1)−2−1のウェルをよくピペッティングし、細胞数をカウントした。
(1)−3−1のウェルをよくピペッティングし、細胞数をカウントした。
(5)G−Rex 24ウェルをよくピペッティングし、1mM EDTA/PBSを使用して細胞を回収し細胞数をカウントした。
・Day 21
The wells in (1)-2-1 were thoroughly pipetted, and the number of cells was counted.
The wells in (1)-3-1 were thoroughly pipetted, and the number of cells was counted.
(5) The G-Rex 24-well plate was thoroughly pipetted, and the cells were recovered using 1 mM EDTA/PBS, and the cell number was counted.
・Day24
(1)−1のウェルをよくピペッティングし、細胞数をカウントした。
(1)−2、(1)−3のウェルをよくピペッティングし、細胞数をカウントした。
・Day 24
(1) The wells in -1 were thoroughly pipetted, and the number of cells was counted.
The cells in wells (1)-2 and (1)-3 were thoroughly pipetted, and the number of cells was counted.
また、生細胞数をカウントし、2x108cellsを5mLのHSC−BANKER(ZENOAQ、CB071)で懸濁し、−80℃で凍結した。凍結細胞を37℃、ウォーターバスにて解凍したのち、Plasma−Lyte Aにて10倍希釈した。解凍した細胞を用いて、実施例4と同様の方法により、K562細胞またはRaji細胞に対する細胞傷害活性を測定した。細胞傷害活性率は、以下のようにA~Dで評価される。評価がC以上である場合に、高い細胞傷害活性が得られ、目的が達成されたと判断される。
A:70%以上
B:60%以上
C:50%以上
D:50%未満
The number of viable cells was counted, and 2x108 cells were suspended in 5mL of HSC-BANKER (ZENOAQ, CB071) and frozen at -80°C. The frozen cells were thawed in a water bath at 37°C, and then diluted 10-fold with Plasma-Lyte A. Using the thawed cells, the cytotoxic activity against K562 cells or Raji cells was measured by the same method as in Example 4. The cytotoxic activity rate is evaluated as A to D as follows. When the evaluation is C or higher, it is determined that high cytotoxic activity is obtained and the purpose is achieved.
A: 70% or more B: 60% or more C: 50% or more D: Less than 50%
結果を図11に示す。E(エフェクター細胞(回収した細胞)):T(標的細胞(K562細胞))=1:1または5:1のいずれの場合においても、回収した細胞集団によるK562細胞およびRaji細胞に対する細胞傷害活性率の評価はAであり、いずれの細胞に対しても高い細胞傷害活性が認められた。 The results are shown in Figure 11. In either case where the ratio of E (effector cells (recovered cells)):T (target cells (K562 cells)) was 1:1 or 5:1, the cytotoxic activity rate of the recovered cell population against K562 cells and Raji cells was evaluated as A, and high cytotoxic activity was observed against both types of cells.
G).実施例7(ドナーMixC,シングルドナーA)
・Day0
凍結保存PBMCを所定の方法で解凍し、以下の5群に調製した細胞懸濁液を6ウェルプレートに1.8mL播種し培養を開始した。
G). Example 7 (Donor Mix C, Single Donor A)
・Day 0
Cryopreserved PBMCs were thawed by a prescribed method, and 1.8 mL of the cell suspension prepared for the following five groups was seeded onto a 6-well plate to initiate culture.
(1)PBMC(CD3−,CD34−)4x105cells/mL,KBM501培地※4,Urelumab[1μg/mL],IL−21[1ng/mL]
(2)PBMC(CD3−,CD34−)4x105cells/mL,NTI培地※1,Urelumab[1μg/mL],IL−21[1ng/mL]
(3)PBMC(CD3−,CD34−)4x105cells/mL,シムレクト、プログラフ入KBM501培地※5,Urelumab[1μg/mL],IL−21[1ng/mL]
(4)PBMC(CD3−,CD34−)4x105cells/mL,シムレクト、プログラフ入NTI培地※6,Urelumab[1μg/mL],IL−21[1ng/mL]
(5)PBMC(CD3+,CD34+)4x105cells/mL,シムレクト、プログラフ入NTI培地※6,Urelumab[1μg/mL],IL−21[1ng/mL]
(1) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, KBM501 medium *4 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(2) PBMC (CD3 − , CD34 − ) 4x10 5 cells/mL, NTI medium *1 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
(3) PBMC ( CD3- , CD34- ) 4x105 cells/mL, Simulect, KBM501 medium containing Prograf *5 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(4) PBMC ( CD3- , CD34- ) 4x105 cells/mL, Simulect, NTI medium containing Prograf *6 , Urelumab [1μg/mL], IL-21 [1ng/mL]
(5) PBMC (CD3 + , CD34 + ) 4 x 10 5 cells/mL, Simulect, NTI medium containing Prograf *6 , Urelumab [1 μg/mL], IL-21 [1 ng/mL]
・Day7
(1)から(5)の各ウェルをよくピペッティングし、細胞数をカウントした。さらに以下のそれぞれの条件の下6ウェルプレートに継代培養した。
(1)−1 新しいKBM501培地※2で5x105cells/3mLにメスアップし、Urelumab[1μg/mL],IL−21[1ng/mL]を添加して播種
(1)−2 新しいKBM501培地※4で5x105cells/3mLにメスアップし播種
(1)−3 新しいKBM501培地※4で5x105cells/1.83mLにメスアップし播種
(1)−4 5x105cellsを分取し、500g,5分遠心後新しいUrelumab[1μg/mL],IL−21[1ng/mL]を添加したKBM501培地※43mLでよく懸濁し播種
Day 7
Each well from (1) to (5) was thoroughly pipetted, and the number of cells was counted. The cells were then subcultured in 6-well plates under each of the following conditions.
(1)-1: Dilute to 5x105 cells/3mL with fresh KBM501 medium *2 , add Urelumab [1μg/mL] and IL-21 [1ng/mL] and sow. (1)-2: Dilute to 5x105 cells/3mL with fresh KBM501 medium *4 and sow. (1)-3: Dilute to 5x105 cells/1.83mL with fresh KBM501 medium * 4 and sow. (1)-4: Take 5x105 cells, centrifuge at 500g for 5 minutes, then suspend in 3mL of fresh KBM501 medium *4 containing Urelumab [1μg/mL] and IL-21 [1ng/mL] and sow.
(2)−1 新しいNTI培地※4で5x105cells/3mLにメスアップし、Urelumab[1μg/mL],IL−21[1ng/mL]を添加して播種
(2)−2 新しいNTI培地※1で5x105cells/3mLにメスアップし播種
(2)−3 新しいNTI培地※1で5x105cells/1.83mLにメスアップし播種
(2)−4 5x105cellsを分取し、500g,5分遠心後新しいUrelumab[1μg/mL],IL−21[1ng/mL]を添加したNTI培地※13mLでよく懸濁し播種
(2)-1: Dilute to 5x105 cells/3mL with new NTI medium *4 , add Urelumab [1μg/mL] and IL-21 [1ng/mL] and sow. (2)-2: Dilute to 5x105 cells/3mL with new NTI medium *1 and sow. (2)-3: Dilute to 5x105 cells/1.83mL with new NTI medium *1 and sow. (2)-4: Take 5x105 cells, centrifuge at 500g for 5 minutes, then suspend well in 3mL of new NTI medium *1 containing Urelumab [1μg/mL] and IL-21 [1ng/mL] and sow.
(5)−1 新しいNTI培地※1で5x105cells/3mLにメスアップし、Urelumab[1μg/mL],IL−21[1ng/mL]を添加して播種
(5)−2 新しいNTI培地※1で5x105cells/3mLにメスアップし播種
(5)−3 新しいNTI培地※1で5x105cells/1.83mLにメスアップし播種
(5)−4 5x105cellsを分取し、500g,5分遠心後新しいUrelumab[1μg/mL],IL−21[1ng/mL]を添加したNTI培地※13mLでよく懸濁し播種
(5)-1: Dilute to 5x105 cells/3mL with new NTI medium *1 , add Urelumab [1μg/mL] and IL-21 [1ng/mL] and sow. (5)-2: Dilute to 5x105 cells/3mL with new NTI medium *1 and sow. (5)-3: Dilute to 5x105 cells/1.83mL with new NTI medium *1 and sow. (5)-4: Take 5x105 cells, centrifuge at 500g for 5 minutes, then suspend well in 3mL of new NTI medium *1 containing Urelumab [1μg/mL] and IL-21 [1ng/mL] and sow.
(3)−5 新しいシムレクト、プログラフ入KBM501培地※52.5mLと細胞懸濁液1.5mLを播種
(4)−5 新しいシムレクト、プログラフ入NTI培地※62.5mLと細胞懸濁液1.5mLを播種
(3)-5 Seed 2.5 mL of new Simulect and Prograf-containing KBM501 medium *5 and 1.5 mL of cell suspension. (4)-5 Seed 2.5 mL of new Simulect and Prograf-containing NTI medium *6 and 1.5 mL of cell suspension.
(1)新しいKBM501培地※43.5mLを入れて継代培養
(2)新しいNTI培地※13.5mLを入れて継代培養
(3)新しいKBM501培地※53.5mLを入れて継代培養
(4)新しいNTI培地※13.5mLを入れて継代培養
(5)新しいNTI培地※13.5mLを入れて継代培養
(1) Add 3.5 mL of new KBM501 medium *4 and subculture . (2) Add 3.5 mL of new NTI medium *1 and subculture. (3) Add 3.5 mL of new KBM501 medium *5 and subculture. (4) Add 3.5 mL of new NTI medium *1 and subculture. (5) Add 3.5 mL of new NTI medium *1 and subculture.
・Day9
Day7で継代培養したすべてのウェルをよくピペッティングし、細胞数をカウントした。(1)−3、(2)−3、(5)−3、(4)−5の各ウェルを5倍希釈(細胞懸濁液0.4mLと新しいメディウム1.6mL)と10倍希釈(細胞懸濁液0.2mLと新しいメディウム1.8mL)で、6ウェルプレートに2mLで継代培養した。
・Day 9
All wells subcultured on Day 7 were thoroughly pipetted and the number of cells was counted. Each of the wells (1)-3, (2)-3, (5)-3, and (4)-5 was subcultured in 2 mL of 5-fold dilution (0.4 mL of cell suspension and 1.6 mL of new medium) and 10-fold dilution (0.2 mL of cell suspension and 1.8 mL of new medium) onto a 6-well plate.
(1)−3−5倍 (1)−3細胞懸濁液0.4mL,KBM501培地※41.6mL
(2)−3−5倍 (2)−3細胞懸濁液0.4mL,NTI培地※11.6mL
(5)−3−5倍 (5)−3細胞懸濁液0.4mL,シムレクト、プログラフ入NTI培地※61.6mL
(4)−5−5倍 (4)−3細胞懸濁液0.4mL,シムレクト、プログラフ入NTI培地※61.6mL
(1)-3-5x (1)-3 cell suspension 0.4mL, KBM501 medium *4 1.6mL
(2)-3-5x (2)-3 Cell suspension 0.4mL, NTI medium *1 1.6mL
(5)-3-5x (5)-3 cell suspension 0.4mL, Simulect, Prograf NTI medium *6 1.6mL
(4)-5-5 times (4)-3 Cell suspension 0.4 mL, Simulect, Prograf-containing NTI medium *6 1.6 mL
(1)−3−10倍 (1)−3細胞懸濁液0.2mL,KBM501培地※41.8mL
(2)−3−10倍 (2)−3細胞懸濁液0.2mL,NTI培地※11.8mL
(5)−3−10倍 (5)−3細胞懸濁液0.2mL,シムレクト、プログラフ入NTI培地※61.8mL
(4)−5−10倍 (4)−3細胞懸濁液0.2mL,シムレクト、プログラフ入NTI培地※61.8mL
(1)-3-10x (1)-3 cell suspension 0.2mL, KBM501 medium *4 1.8mL
(2)-3-10x (2)-3 Cell suspension 0.2mL, NTI medium *1 1.8mL
(5)-3-10x (5)-3 cell suspension 0.2mL, Simulect, Prograf NTI medium *6 1.8mL
(4)-5-10x (4)-3 Cell suspension 0.2mL, Simulect, Prograf NTI medium *6 1.8mL
・Day11
Day9で継代培養したすべてのウェル、(3)−5、(3)、(4)をよくピペッティングし、細胞数をカウントした。(1)−3−5倍、(2)−3−5倍、(5)−3−5倍、(5)−3−5−10倍のウェルからそれぞれ新しい培地で2x105cells/2mLに調製し6ウェルプレートに継代培養した。
(1)−3−5倍−2E5 細胞懸濁液2x105cells/2mL,KBM501培地※4
(2)−3−5倍−2E5 細胞懸濁液2x105cells/2mL,NTI培地※1
(5)−3−5倍−2E5 細胞懸濁液2x105cells/2mL,シムレクト、プログラフ入NTI培地※6
(5)−3−10倍−2E5 細胞懸濁液2x105cells/2mL,シムレクト、プログラフ入NTI培地※6
・Day 11
All wells subcultured on Day 9, (3)-5, (3), and (4) were thoroughly pipetted and the number of cells was counted. From wells (1)-3-5x, (2)-3-5x, (5)-3-5x, and (5)-3-5-10x, fresh medium was added to each well to prepare 2 x 105 cells/2 mL, and the cells were subcultured in a 6-well plate.
(1)-3-5x-2E5 cell suspension 2x105 cells/2mL, KBM501 medium *4
(2)-3-5x-2E5 cell suspension 2x105 cells/2mL, NTI medium *1
(5)-3-5x-2E5 cell suspension 2x105 cells/2mL, Simulect, Prograf-containing NTI medium *6
(5)-3-10x-2E5 cell suspension 2x105 cells/2mL, Simulect, Prograf-containing NTI medium *6
・Day14
Day11で継代培養したすべてのウェル、(2)−3−5倍、(2)−3−10倍をよくピペッティングし、細胞数をカウントした。(2)−3−5倍、(2)−3−10倍のウェルからそれぞれ新しい培地で4x105cells/2mLに調製し6ウェルプレートに継代培養した。
・Day 14
All wells subcultured on Day 11, (2)-3-5x, and (2)-3-10x were thoroughly pipetted and the number of cells was counted. From the (2)-3-5x and (2)-3-10x wells, fresh medium was added to each well to prepare 4 x 105 cells/2 mL, and the cells were subcultured in a 6-well plate.
結果を図12に示す。 The results are shown in Figure 12.
H).実施例8(ドナーMix)
凍結保存PBMCを37℃のウォーターバスにて完全に解凍し、KBM501/5%UG培地※7で10倍に希釈した。500×g,5分間遠心後、以下に示す6群の培地にて4×105cells/mLに調製し、12ウェルプレートに0.8mL播種し培養を開始した。
H). Example 8 (Donor Mix)
Cryopreserved PBMCs were completely thawed in a 37°C water bath and diluted 10-fold with KBM501/5% UG medium *7 . After centrifugation at 500×g for 5 minutes, the cells were adjusted to 4×10 5 cells/mL with the following 6 groups of medium, and 0.8 mL was seeded on a 12-well plate to start culture.
・Day0
(1)control:シムレクトおよびタクロリムスを含むKBM501/5%UG培地※8
(2)Urelumab:シムレクトおよびタクロリムスを含むKBM501/5%UG培地※8,1ng/mL IL−21,1μg/mL Urelumab
(3)1μg/mL:シムレクトおよびタクロリムスを含むKBM501/5%UG培地※8,1ng/mL IL−21,1μg/mL rhs4−1BBL
(4)5μg/mL:シムレクトおよびタクロリムスを含むKBM501/5%UG培地※8,1ng/mL IL−21,5μg/mL rhs4−1BBL
(5)10μg/mL:シムレクトおよびタクロリムスを含むKBM501/5%UG培地※8,1ng/mL IL−21,10μg/mL rhs4−1BBL
(6)IL−21:シムレクトおよびタクロリムスを含むKBM501/5%UG培地※8,1ng/mL IL−21
rhs4−1BBL=recombinant human soluble 4−1BB(CD137) ligand(配列番号:1のアミノ酸配列からなるポリペプチド)
・Day 0
(1) Control: KBM501/5% UG medium containing Simulect and tacrolimus *8
(2) Urelumab: KBM501/5% UG medium containing Simulect and tacrolimus, 1 ng/mL IL-21, 1 μg/mL Urelumab
(3) 1 μg/mL: KBM501/5% UG medium containing Simulect and tacrolimus, 1 ng/mL IL-21, 1 μg/mL rhs4-1BBL
(4) 5 μg/mL: KBM501/5% UG medium containing Simulect and tacrolimus *8 , 1 ng/mL IL-21, 5 μg/mL rhs4-1BBL
(5) 10 μg/mL: KBM501/5% UG medium containing Simulect and tacrolimus *8 , 1 ng/mL IL-21, 10 μg/mL rhs4-1BBL
(6) IL-21: KBM501/5% UG medium containing Simulect and tacrolimus, 1 ng/mL IL-21
rhs4-1BBL = recombinant human soluble 4-1BB (CD137) ligand (polypeptide consisting of the amino acid sequence of SEQ ID NO: 1)
・Day7
(2)~(5)にKBM501/5%UG培地を800μL追加した。
Day 7
800 μL of KBM501/5% UG medium was added to (2) to (5).
・Day9
(1)、(6)にそれぞれKBM501/5%UG培地を1.6mL追加した。(2)~(5)は各群、ウェルをよくピペッティングし細胞数をカウントした。その後、ウェルから細胞懸濁液を分取、2×105cells/mLとなるようKBM501培地/5%UGにて調製し、6ウェルプレートに継代した。
・Day 9
1.6 mL of KBM501/5% UG medium was added to each of (1) and (6). For (2) to (5), the wells were thoroughly pipetted and the number of cells was counted. Then, a cell suspension was taken from the well, adjusted to 2 x 10 5 cells/mL in KBM501 medium/5% UG, and subcultured in a 6-well plate.
・Day11
(6)およびDay9で6ウェルプレートに継代した(2)~(5)について各群、ウェルをよくピペッティングし細胞数をカウントした。(3)はそのまま培養を維持し、そのほかの群はウェルから細胞懸濁液を分取、(2)、(4)、(5)は2.35×105cells/mL、(6)は1.84×105cells/mLとなるようKBM501培地/5%UGにて調製し、6ウェルプレートに継代した。
・Day 11
For (6) and (2) to (5) that were passaged to a 6-well plate on Day 9, the wells of each group were thoroughly pipetted and the number of cells was counted. (3) was maintained in culture as is, and for the other groups, cell suspensions were taken from the wells and adjusted to 2.35 x 105 cells/mL for (2), (4), and (5), and 1.84 x 105 cells/mL for (6) in KBM501 medium/5% UG, and passaged to a 6-well plate.
・Day14
全群(Day0で12ウェルプレートに播種した(1)、Day9あるいはDay11で6ウェルプレートに継代した(2)~(6))、1mM EDTA/PBSを使用して細胞を回収し細胞数をカウントした。また、回収した細胞のうち1×105cellsを以下に示す抗体(各抗体濃度1μg/mL)にて染色した。
・Day 14
For all groups ((1) seeded onto 12-well plates on Day 0, (2) to (6) passaged onto 6-well plates on Day 9 or Day 11), cells were harvested using 1 mM EDTA/PBS and the cell numbers were counted. In addition, 1 x 10 5 cells out of the harvested cells were stained with the following antibodies (each antibody concentration 1 μg/mL).
4℃、30分間染色後、遠心分離(500×g,5分間)、上清を除去し、PBSに懸濁したのち、フローサイトメーター(BD LSRFortessa,BDバイオサイエンス社)を用いて測定を行い、FlowJoソフトウェア(FLOWJO,LLC)で解析した。 After staining at 4°C for 30 minutes, the cells were centrifuged (500 x g, 5 minutes), the supernatant was removed, and the cells were suspended in PBS. Measurements were then performed using a flow cytometer (BD LSRFortessa, BD Biosciences) and analyzed using FlowJo software (FLOWJO, LLC).
細胞集団の増幅変化の結果を図13に示す。各細胞表面マーカーについてのフローサイトメトリーの結果を図14および図15に示す。 The results of the amplification changes in the cell population are shown in Figure 13. The flow cytometry results for each cell surface marker are shown in Figures 14 and 15.
rhs−4−1BBLを用いた場合においても、(1)control群(rhs−4−1BBLまたはanti 4−1BBもIL−21も添加しない場合)および(6)IL−21群(rhs−4−1BBLまたはanti 4−1BBを添加しないがIL−21を添加する場合)と比較して、効率的な細胞の増殖が得られた(図13)。 Even when rhs-4-1BBL was used, efficient cell proliferation was obtained compared to (1) the control group (when neither rhs-4-1BBL, anti-4-1BB, nor IL-21 was added) and (6) the IL-21 group (when rhs-4-1BBL or anti-4-1BB was not added but IL-21 was added) (Figure 13).
※1:5% UltraGRO(AventaCell、HPCPLCRL10)および2U/mLヘパリンナトリウム(ニプロ)を添加したNTI(FUKOKU,T2108251)
※2:5% UltraGRO(AventaCell、HPCPLCRL10)、2U/mLヘパリンナトリウム(ニプロ)および100Units/mLイムネースを添加したRPMI1640(ナカライ,30264−85)
※3:5% UltraGRO(AventaCell、HPCPLCRL10)および2U/mLヘパリンナトリウム(ニプロ)を添加したKBM502(KOHJIN BIO,16025020)
※4:5% UltraGRO(AventaCell、HPCPLCRL10)および2U/mLヘパリンナトリウム(ニプロ)を添加したKBM501(KOHJIN BIO,16025015)
※5:5% UltraGRO(AventaCell、HPCPLCRL10)および2U/mLヘパリンナトリウム(ニプロ)、シムレクト0.5μg/mL、プログラフ1ng/mLを添加したKBM501(KOHJIN BIO,16025015)
※6:5% UltraGRO(AventaCell、HPCPLCRL10)および2U/mLヘパリンナトリウム(ニプロ)、シムレクト0.5μg/mL、プログラフ1ng/mLを添加したNTI(FUKOKU,T2108251)
*1: NTI (FUKOKU, T2108251) supplemented with 5% UltraGRO (AventaCell, HPCPLCRL10) and 2 U/mL heparin sodium (Nipro)
*2: RPMI1640 (Nacalai, 30264-85) supplemented with 5% UltraGRO (AventaCell, HPCPLCRL10), 2U/mL heparin sodium (Nipro), and 100Units/mL Immunase
*3: KBM502 (KOHJIN BIO, 16025020) supplemented with 5% UltraGRO (AventaCell, HPCPLCRL10) and 2U/mL heparin sodium (Nipro)
*4: KBM501 (KOHJIN BIO, 16025015) supplemented with 5% UltraGRO (AventaCell, HPCPLCRL10) and 2U/mL heparin sodium (Nipro)
*5: KBM501 (KOHJIN BIO, 16025015) supplemented with 5% UltraGRO (AventaCell, HPCPLCRL10) and 2U/mL heparin sodium (Nipro), 0.5μg/mL Simulect, and 1ng/mL Prograf.
*6: NTI (FUKOKU, T2108251) containing 5% UltraGRO (AventaCell, HPCPLCRL10) and 2U/mL heparin sodium (Nipro), Simulect 0.5μg/mL, and Prograf 1ng/mL
※7:5% UltraGROおよび2U/mLヘパリンナトリウムを添加したKBM 501
※8:5% UltraGRO、2U/mLヘパリンナトリウム、0.5μg/mLシムレクト、1ng/mLプログラフを添加したKBM 501
*7: KBM 501 supplemented with 5% UltraGRO and 2 U/mL heparin sodium
*8: KBM 501 supplemented with 5% UltraGRO, 2 U/mL heparin sodium, 0.5 μg/mL Simulect, and 1 ng/mL Prograf.
[使用した試薬および器具について]
・RPMI1640(ナカライテスク,30264−85)
・KBM501(KOHJIN BIO,16025015)(IL−2 2810IU/mL含有)
・KBM502(KOHJIN BIO,16025020)(IL−2 281IU/mL含有)
・NTI(FUKOKU)
・CTS Immune Cell SR(gibco,A25961−01)
・UltraGRO(AventaCell,HPCPLCRL10)
・FBS(NICHIREI,174012,19C00A)
・ヘパリンナトリウム(ニプロ)
・Urelumab(Urelumab)(Creative Biolabs,TAB−179)
・IL−21(PEPROTECH,AF−200−21−10μg)
・IL−18(Biolegend,592102)
・イムネース(塩野義製薬)
・シムレクト(NOVARTIS)
・プログラフ(astellas)
・ペニシリン−ストレプトマイシン混合溶液(ナカライテスク,26253−84)
・0.5M EDTA(invitrogen,15575−038)
・PBS(ナカライテスク,14249−24)
・7−AAD(Beckman Coulter,A07704)
・APC anti−human CD107a Antibody(Biolegend,328620)
・APC Mouse IgG1,κIsotype Ctrl Antibody(Biolegend,400120)
・EasySep Human NK Cell Enrichment Kit (STEMCELL Technologies,19055)
・6ウェルプレート(Termo,140675)
・12ウェルプレート(Termo,150628)
・24ウェルプレート(Thermo,142475)
・G−Rex 24ウェルプレート(BMBio,BMA−GP1024)
・T75フラスコ(TermoFisher,156499)
・T225フラスコ(TermoFisher,159933)
・10cm ディッシュ(Falcon,3530003)
・96ウェルプレート(IWAKI,4870−800SP)
・EZSPHERE(登録商標)6ウェルプレート(IWAKI,4810−900SP)
[Reagents and equipment used]
RPMI1640 (Nacalai Tesque, 30264-85)
・KBM501 (KOHJIN BIO, 16025015) (contains IL-2 2810IU/mL)
・KBM502 (KOHJIN BIO, 16025020) (contains IL-2 281IU/mL)
・NTI (FUKOKU)
・CTS Immune Cell SR (gibco, A25961-01)
・UltraGRO (AventaCell, HPCPLCRL10)
・FBS (NICHIREI, 174012, 19C00A)
・Heparin sodium (Nipro)
・Urelumab (Creative Biolabs, TAB-179)
・IL-21 (PEPROTECH, AF-200-21-10μg)
・IL-18 (Biolegend, 592102)
・Immunace (Shionogi & Co., Ltd.)
・Simrect (NOVARTIS)
・Prograf (Astellas)
Penicillin-streptomycin mixed solution (Nacalai Tesque, 26253-84)
・0.5M EDTA (invitrogen, 15575-038)
PBS (Nacalai Tesque, 14249-24)
・7-AAD (Beckman Coulter, A07704)
・APC anti-human CD107a Antibody (Biolegend, 328620)
・APC Mouse IgG1, κIsotype Ctrl Antibody (Biolegend, 400120)
・EasySep Human NK Cell Enrichment Kit (STEMCELL Technologies, 19055)
・6-well plate (Termo, 140675)
・12-well plate (Termo, 150628)
・24-well plate (Thermo, 142475)
G-Rex 24-well plate (BMBio, BMA-GP1024)
- T75 flask (ThermoFisher, 156499)
- T225 flask (ThermoFisher, 159933)
・10cm dish (Falcon, 3530003)
・96-well plate (IWAKI, 4870-800SP)
EZSPHERE (registered trademark) 6-well plate (IWAKI, 4810-900SP)
・Alexa Fluor(登録商標)700標識抗ヒトCD56抗体(Biolegend,318316)
・APC標識抗ヒトNKp30抗体(Biolegend,325210)
・PerCP/Cy5.5標識抗ヒトCD3抗体(Biolegend,300430)
・PE−Cy7標識抗ヒトCD16抗体(Biolegend,302016)
・PE標識抗ヒトCD19抗体(Biolegend,302208)
・FITC標識抗ヒトCD14抗体(Biolegend,325604)
・APC−Cy7標識抗ヒトCD34抗体(Biolegend,343514)
Alexa Fluor® 700-labeled anti-human CD56 antibody (Biolegend, 318316)
・APC-labeled anti-human NKp30 antibody (Biolegend, 325210)
PerCP/Cy5.5-labeled anti-human CD3 antibody (Biolegend, 300430)
PE-Cy7-labeled anti-human CD16 antibody (Biolegend, 302016)
PE-labeled anti-human CD19 antibody (Biolegend, 302208)
FITC-labeled anti-human CD14 antibody (Biolegend, 325604)
APC-Cy7-labeled anti-human CD34 antibody (Biolegend, 343514)
・FITC標識抗ヒトCD49a抗体(Biolegend,328307)
・PE標識抗ヒトCD49c抗体(Biolegend,343803)
・APC標識抗ヒトCD61抗体(Biolegend,336412)
・PE−Dazzle594標識抗ヒトNKp30抗体(Biolegend,325231)
・PE−Cy7標識抗ヒトCCR5抗体(Biolegend,359107)
・Pacific BlueTM標識抗ヒトCCR6抗体(Biolegend,353438)
・APC−Cy7標識抗ヒトCXCR3抗体(Biolegend,353721)
FITC-labeled anti-human CD49a antibody (Biolegend, 328307)
PE-labeled anti-human CD49c antibody (Biolegend, 343803)
APC-labeled anti-human CD61 antibody (Biolegend, 336412)
PE-Dazzle594-labeled anti-human NKp30 antibody (Biolegend, 325231)
PE-Cy7-labeled anti-human CCR5 antibody (Biolegend, 359107)
Pacific Blue™-labeled anti-human CCR6 antibody (Biolegend, 353438)
APC-Cy7-labeled anti-human CXCR3 antibody (Biolegend, 353721)
・KBM 501(コージンバイオ)
・UltraGRO(AventaCell、HPCPLCRL10)
・ヘパリンナトリウム(ニプロ)
・シムレクト(ノバルティスファーマ)
・プログラフ(アステラス製薬)
・Recombinant Human 4−1BB Ligand Animal−Free manufactured(PREPROTECH)
・KBM 501 (Kohjin Bio)
・UltraGRO (AventaCell, HPCPLCRL10)
・Heparin sodium (Nipro)
・Simlect (Novartis Pharma)
・Prograf (Astellas Pharma)
・Recombinant Human 4-1BB Ligand Animal-Free manufactured (PREPROTECH)
[配列表に記載した配列について]
配列番号:1 Recombinant Human 4−1BB Ligand Animal−Free manufactured(PREPROTECH)
配列番号:2 Recombinant Human 4−1BB Ligand(N−Fc)#AP76391(Signalway Antibody LLC)
配列番号:3 Extracellular domain of 4−1BBL
[Sequences listed in the sequence listing]
SEQ ID NO: 1 Recombinant Human 4-1BB Ligand Animal-Free manufactured (PREPROTECH)
SEQ ID NO: 2 Recombinant Human 4-1BB Ligand (N-Fc) #AP76391 (Signalway Antibody LLC)
SEQ ID NO: 3 Extracellular domain of 4-1BBL
Claims (6)
a)単核球を含む細胞集団からCD3陽性細胞およびCD34陽性細胞を除去した細胞集団を、IL−2、IL−21および4−1BBアゴニストを含む培地中で1日以上培養する工程であって、IL−21および4−1BBアゴニストが液性因子である、工程。 A method for producing a cell population containing CD3-negative and CD56-positive cells, comprising the steps of:
a) culturing a cell population obtained by removing CD3-positive cells and CD34-positive cells from a cell population containing mononuclear cells in a medium containing IL-2, IL-21 and a 4-1BB agonist for one day or more, wherein the IL-21 and the 4-1BB agonist are humoral factors.
b)工程a)で培養した細胞集団を、IL−2および4−1BBアゴニストを含み、IL−21を含まない培地で継代し、1日以上培養する工程。 The method of claim 1 further comprising the steps of:
b) Passaging the cell population cultured in step a) in a medium containing IL-2 and a 4-1BB agonist but not containing IL-21, and culturing the medium for one day or more.
c)工程b)で培養した細胞集団を、IL−2を含む培地で継代し、1日以上培養する工程。 The method of claim 2 further comprising the steps of:
c) Passaging the cell population cultured in step b) in a medium containing IL-2 and culturing for one day or more.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2023-119426 | 2023-07-21 | ||
JP2023119426 | 2023-07-21 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2025022372A1 true WO2025022372A1 (en) | 2025-01-30 |
Family
ID=94374126
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/IB2024/059123 WO2025022372A1 (en) | 2023-07-21 | 2024-09-20 | Cell population production method |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2025022372A1 (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2013027385A (en) * | 2011-06-24 | 2013-02-07 | Kyushu Univ | Method for amplifying nk cells |
JP2021534835A (en) * | 2018-08-30 | 2021-12-16 | エイチシーダブリュー バイオロジックス インコーポレイテッド | Single-chain and multi-chain chimeric polypeptides and how to use them |
JP2022520871A (en) * | 2019-02-20 | 2022-04-01 | ラトガーズ、ザ ステイト ユニバーシティ オブ ニュージャージー | Expansion of natural killer cells and chimeric antigen receptor-modified cells |
JP2022548861A (en) * | 2019-09-13 | 2022-11-22 | リサーチ インスティチュート アット ネイションワイド チルドレンズ ホスピタル | Universal Donor Selection Method for Identifying NK Cell Donors |
-
2024
- 2024-09-20 WO PCT/IB2024/059123 patent/WO2025022372A1/en unknown
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2013027385A (en) * | 2011-06-24 | 2013-02-07 | Kyushu Univ | Method for amplifying nk cells |
JP2021534835A (en) * | 2018-08-30 | 2021-12-16 | エイチシーダブリュー バイオロジックス インコーポレイテッド | Single-chain and multi-chain chimeric polypeptides and how to use them |
JP2022520871A (en) * | 2019-02-20 | 2022-04-01 | ラトガーズ、ザ ステイト ユニバーシティ オブ ニュージャージー | Expansion of natural killer cells and chimeric antigen receptor-modified cells |
JP2022548861A (en) * | 2019-09-13 | 2022-11-22 | リサーチ インスティチュート アット ネイションワイド チルドレンズ ホスピタル | Universal Donor Selection Method for Identifying NK Cell Donors |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP6799895B2 (en) | Production method of TCRγδ + T cells | |
JP5156382B2 (en) | Method for producing T cell population | |
JP7604000B2 (en) | Methods for generating modified human primary blood dendritic cell lines | |
ES2771248T3 (en) | Differentiation of ex vivo NK cells from CD34 + hematopoietic cells | |
US20240254446A1 (en) | Population of CD3-negative cells that express chemokine receptor and cell adhesion molecule, use of the same, and method for producing the same | |
US20220411754A1 (en) | Methods and compositions for enhanced expansion and cytotoxicity of natural killer cells | |
JP2019502725A (en) | Compositions and methods for immune cell regulation in adoptive immunotherapy | |
CN107922925A (en) | Method for natural killer cells amplification | |
EP3000876B1 (en) | Method for preparing nk cells | |
US20230265390A1 (en) | Enhanced expansion and cytotoxicity of engineered natural killer cells and uses thereof | |
JP2022542321A (en) | NK cell compositions and preparations for immunotherapy and methods for their production | |
US20170065690A1 (en) | Tscm CELLS AND METHODS FOR USE | |
JP2018531022A6 (en) | Methods for generating modified human primary blood dendritic cell lines | |
JP2024045306A (en) | Methods for producing and using embryonic mesenchymal progenitor cells | |
CN113613723A (en) | Method for producing cell population containing NK cells | |
AU2019381526B2 (en) | Method for culturing cord blood-derived natural killer cells using transformed T-cells | |
JP2022540267A (en) | CIML NK cells and methods therefor | |
WO2025022372A1 (en) | Cell population production method | |
JP2023504075A (en) | Method for obtaining CAR-NK cells | |
HK40024937A (en) | Methods for the production of tcr gamma delta+ t cells | |
HK1245831B (en) | Methods for the production of tcr gamma delta+ t cells | |
JP2010094123A (en) | Method for producing lymphocyte |
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: 24844991 Country of ref document: EP Kind code of ref document: A1 |