WO2007040105A1 - T細胞集団の製造方法 - Google Patents
T細胞集団の製造方法 Download PDFInfo
- Publication number
- WO2007040105A1 WO2007040105A1 PCT/JP2006/319105 JP2006319105W WO2007040105A1 WO 2007040105 A1 WO2007040105 A1 WO 2007040105A1 JP 2006319105 W JP2006319105 W JP 2006319105W WO 2007040105 A1 WO2007040105 A1 WO 2007040105A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- culture
- cell population
- cell
- group
- Prior art date
Links
- 238000004519 manufacturing process Methods 0.000 title claims abstract description 79
- 210000004027 cell Anatomy 0.000 claims abstract description 524
- 210000001744 T-lymphocyte Anatomy 0.000 claims abstract description 394
- 239000012634 fragment Substances 0.000 claims abstract description 120
- 102100036301 C-C chemokine receptor type 7 Human genes 0.000 claims abstract description 119
- 101000716065 Homo sapiens C-C chemokine receptor type 7 Proteins 0.000 claims abstract description 119
- 238000000034 method Methods 0.000 claims abstract description 115
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 claims abstract description 79
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 claims abstract description 79
- 108010067306 Fibronectins Proteins 0.000 claims abstract description 76
- 102000016359 Fibronectins Human genes 0.000 claims abstract description 76
- 101001018097 Homo sapiens L-selectin Proteins 0.000 claims abstract description 76
- 102100033467 L-selectin Human genes 0.000 claims abstract description 76
- 102100027207 CD27 antigen Human genes 0.000 claims abstract description 56
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 claims abstract description 56
- 239000000203 mixture Substances 0.000 claims abstract description 28
- 239000000427 antigen Substances 0.000 claims description 132
- 102000036639 antigens Human genes 0.000 claims description 132
- 108091007433 antigens Proteins 0.000 claims description 132
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 72
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 claims description 70
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 claims description 70
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 63
- 108090000623 proteins and genes Proteins 0.000 claims description 53
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 48
- 229920001184 polypeptide Polymers 0.000 claims description 46
- 239000003814 drug Substances 0.000 claims description 45
- 239000003446 ligand Substances 0.000 claims description 42
- 238000002360 preparation method Methods 0.000 claims description 35
- 239000004480 active ingredient Substances 0.000 claims description 34
- 239000013598 vector Substances 0.000 claims description 30
- 230000004936 stimulating effect Effects 0.000 claims description 26
- 238000012258 culturing Methods 0.000 claims description 19
- 201000010099 disease Diseases 0.000 claims description 19
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 19
- 108010012236 Chemokines Proteins 0.000 claims description 16
- 102000019034 Chemokines Human genes 0.000 claims description 16
- 150000001413 amino acids Chemical class 0.000 claims description 15
- 241000700605 Viruses Species 0.000 claims description 9
- 238000009472 formulation Methods 0.000 claims description 5
- 241000702421 Dependoparvovirus Species 0.000 claims description 4
- 230000001177 retroviral effect Effects 0.000 claims 1
- 239000000126 substance Substances 0.000 abstract description 8
- 230000001472 cytotoxic effect Effects 0.000 description 76
- 241000699666 Mus <mouse, genus> Species 0.000 description 75
- 108010002350 Interleukin-2 Proteins 0.000 description 54
- 102000000588 Interleukin-2 Human genes 0.000 description 54
- 238000004113 cell culture Methods 0.000 description 43
- 239000002609 medium Substances 0.000 description 40
- 238000005259 measurement Methods 0.000 description 36
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 34
- 239000000243 solution Substances 0.000 description 32
- 230000027455 binding Effects 0.000 description 30
- 230000006698 induction Effects 0.000 description 30
- 210000004748 cultured cell Anatomy 0.000 description 29
- 230000000694 effects Effects 0.000 description 28
- 239000002953 phosphate buffered saline Substances 0.000 description 28
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 description 25
- 230000000638 stimulation Effects 0.000 description 25
- 239000012980 RPMI-1640 medium Substances 0.000 description 21
- 235000001014 amino acid Nutrition 0.000 description 21
- 230000000735 allogeneic effect Effects 0.000 description 19
- 101000578784 Homo sapiens Melanoma antigen recognized by T-cells 1 Proteins 0.000 description 18
- 102100028389 Melanoma antigen recognized by T-cells 1 Human genes 0.000 description 18
- 238000004458 analytical method Methods 0.000 description 18
- 238000002474 experimental method Methods 0.000 description 18
- 210000004698 lymphocyte Anatomy 0.000 description 18
- 206010036790 Productive cough Diseases 0.000 description 17
- 239000013612 plasmid Substances 0.000 description 17
- 210000003802 sputum Anatomy 0.000 description 17
- 208000024794 sputum Diseases 0.000 description 17
- 239000000306 component Substances 0.000 description 16
- 210000002381 plasma Anatomy 0.000 description 16
- 206010028980 Neoplasm Diseases 0.000 description 15
- 230000000259 anti-tumor effect Effects 0.000 description 15
- 229940024606 amino acid Drugs 0.000 description 14
- 210000002966 serum Anatomy 0.000 description 14
- 238000010186 staining Methods 0.000 description 14
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 13
- 239000012228 culture supernatant Substances 0.000 description 13
- 238000006467 substitution reaction Methods 0.000 description 13
- VZSRBBMJRBPUNF-UHFFFAOYSA-N 2-(2,3-dihydro-1H-inden-2-ylamino)-N-[3-oxo-3-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)propyl]pyrimidine-5-carboxamide Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C(=O)NCCC(N1CC2=C(CC1)NN=N2)=O VZSRBBMJRBPUNF-UHFFFAOYSA-N 0.000 description 12
- 230000006870 function Effects 0.000 description 12
- 238000009169 immunotherapy Methods 0.000 description 12
- 238000012546 transfer Methods 0.000 description 12
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 11
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 11
- DEGAKNSWVGKMLS-UHFFFAOYSA-N calcein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC(CN(CC(O)=O)CC(O)=O)=C(O)C=C1OC1=C2C=C(CN(CC(O)=O)CC(=O)O)C(O)=C1 DEGAKNSWVGKMLS-UHFFFAOYSA-N 0.000 description 11
- 229960002378 oftasceine Drugs 0.000 description 11
- 210000000952 spleen Anatomy 0.000 description 11
- 241000588724 Escherichia coli Species 0.000 description 10
- 241000699670 Mus sp. Species 0.000 description 10
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 10
- 210000004369 blood Anatomy 0.000 description 10
- 239000008280 blood Substances 0.000 description 10
- 229940079593 drug Drugs 0.000 description 10
- 238000011534 incubation Methods 0.000 description 10
- -1 CH-271 Chemical compound 0.000 description 9
- 238000007792 addition Methods 0.000 description 9
- 210000000612 antigen-presenting cell Anatomy 0.000 description 9
- 230000036961 partial effect Effects 0.000 description 9
- 102000004169 proteins and genes Human genes 0.000 description 9
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 8
- 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 8
- 239000007789 gas Substances 0.000 description 8
- 229960002897 heparin Drugs 0.000 description 8
- 229920000669 heparin Polymers 0.000 description 8
- 238000003780 insertion Methods 0.000 description 8
- 230000037431 insertion Effects 0.000 description 8
- 239000000047 product Substances 0.000 description 8
- 235000018102 proteins Nutrition 0.000 description 8
- 229940124597 therapeutic agent Drugs 0.000 description 8
- 239000006285 cell suspension Substances 0.000 description 7
- 238000012217 deletion Methods 0.000 description 7
- 230000037430 deletion Effects 0.000 description 7
- 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 7
- 210000000265 leukocyte Anatomy 0.000 description 7
- 239000007790 solid phase Substances 0.000 description 7
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 6
- 102100037850 Interferon gamma Human genes 0.000 description 6
- 108010074328 Interferon-gamma Proteins 0.000 description 6
- 108010065805 Interleukin-12 Proteins 0.000 description 6
- 102000013462 Interleukin-12 Human genes 0.000 description 6
- 230000000961 alloantigen Effects 0.000 description 6
- 210000003969 blast cell Anatomy 0.000 description 6
- 230000010261 cell growth Effects 0.000 description 6
- 239000012636 effector Substances 0.000 description 6
- 238000000684 flow cytometry Methods 0.000 description 6
- 210000003071 memory t lymphocyte Anatomy 0.000 description 6
- 238000000926 separation method Methods 0.000 description 6
- 238000007447 staining method Methods 0.000 description 6
- 230000001225 therapeutic effect Effects 0.000 description 6
- 241000239290 Araneae Species 0.000 description 5
- 102100036846 C-C motif chemokine 21 Human genes 0.000 description 5
- 238000002965 ELISA Methods 0.000 description 5
- 101000713085 Homo sapiens C-C motif chemokine 21 Proteins 0.000 description 5
- 239000008351 acetate buffer Substances 0.000 description 5
- BQRGNLJZBFXNCZ-UHFFFAOYSA-N calcein am Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC(CN(CC(=O)OCOC(C)=O)CC(=O)OCOC(C)=O)=C(OC(C)=O)C=C1OC1=C2C=C(CN(CC(=O)OCOC(C)=O)CC(=O)OCOC(=O)C)C(OC(C)=O)=C1 BQRGNLJZBFXNCZ-UHFFFAOYSA-N 0.000 description 5
- 201000011510 cancer Diseases 0.000 description 5
- 238000006243 chemical reaction Methods 0.000 description 5
- 230000035605 chemotaxis Effects 0.000 description 5
- 239000001963 growth medium Substances 0.000 description 5
- 238000001802 infusion Methods 0.000 description 5
- 201000001441 melanoma Diseases 0.000 description 5
- 239000006228 supernatant Substances 0.000 description 5
- 230000004083 survival effect Effects 0.000 description 5
- 102000004127 Cytokines Human genes 0.000 description 4
- 108090000695 Cytokines Proteins 0.000 description 4
- 108090000978 Interleukin-4 Proteins 0.000 description 4
- 102000004388 Interleukin-4 Human genes 0.000 description 4
- 108010002586 Interleukin-7 Proteins 0.000 description 4
- 230000021164 cell adhesion Effects 0.000 description 4
- 238000011156 evaluation Methods 0.000 description 4
- 230000001965 increasing effect Effects 0.000 description 4
- 230000000977 initiatory effect Effects 0.000 description 4
- 239000007788 liquid Substances 0.000 description 4
- 239000003550 marker Substances 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 229910052757 nitrogen Inorganic materials 0.000 description 4
- 150000007523 nucleic acids Chemical group 0.000 description 4
- 229920001778 nylon Polymers 0.000 description 4
- 230000008569 process Effects 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 4
- 239000011550 stock solution Substances 0.000 description 4
- 238000003860 storage Methods 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- 108020004414 DNA Proteins 0.000 description 3
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 3
- 241000713666 Lentivirus Species 0.000 description 3
- 101710160107 Outer membrane protein A Proteins 0.000 description 3
- 239000012979 RPMI medium Substances 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 239000002458 cell surface marker Substances 0.000 description 3
- 238000002659 cell therapy Methods 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 230000008859 change Effects 0.000 description 3
- 230000004940 costimulation Effects 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 239000012895 dilution Substances 0.000 description 3
- 239000012530 fluid Substances 0.000 description 3
- 238000001415 gene therapy Methods 0.000 description 3
- 239000011521 glass Substances 0.000 description 3
- 208000024908 graft versus host disease Diseases 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 230000004073 interleukin-2 production Effects 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 210000001165 lymph node Anatomy 0.000 description 3
- 229930182817 methionine Natural products 0.000 description 3
- 210000000822 natural killer cell Anatomy 0.000 description 3
- 239000013642 negative control Substances 0.000 description 3
- 108020004707 nucleic acids Proteins 0.000 description 3
- 102000039446 nucleic acids Human genes 0.000 description 3
- 210000005105 peripheral blood lymphocyte Anatomy 0.000 description 3
- 230000035945 sensitivity Effects 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 210000004881 tumor cell Anatomy 0.000 description 3
- 241000701161 unidentified adenovirus Species 0.000 description 3
- 241001430294 unidentified retrovirus Species 0.000 description 3
- 239000013603 viral vector Substances 0.000 description 3
- 206010003445 Ascites Diseases 0.000 description 2
- 241000894006 Bacteria Species 0.000 description 2
- OBMZMSLWNNWEJA-XNCRXQDQSA-N C1=CC=2C(C[C@@H]3NC(=O)[C@@H](NC(=O)[C@H](NC(=O)N(CC#CCN(CCCC[C@H](NC(=O)[C@@H](CC4=CC=CC=C4)NC3=O)C(=O)N)CC=C)NC(=O)[C@@H](N)C)CC3=CNC4=C3C=CC=C4)C)=CNC=2C=C1 Chemical compound C1=CC=2C(C[C@@H]3NC(=O)[C@@H](NC(=O)[C@H](NC(=O)N(CC#CCN(CCCC[C@H](NC(=O)[C@@H](CC4=CC=CC=C4)NC3=O)C(=O)N)CC=C)NC(=O)[C@@H](N)C)CC3=CNC4=C3C=CC=C4)C)=CNC=2C=C1 OBMZMSLWNNWEJA-XNCRXQDQSA-N 0.000 description 2
- 108010083702 Chemokine CCL21 Proteins 0.000 description 2
- 102000006435 Chemokine CCL21 Human genes 0.000 description 2
- 102000016911 Deoxyribonucleases Human genes 0.000 description 2
- 108010053770 Deoxyribonucleases Proteins 0.000 description 2
- 206010059866 Drug resistance Diseases 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 230000010556 Heparin Binding Activity Effects 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 2
- 101000815628 Homo sapiens Regulatory-associated protein of mTOR Proteins 0.000 description 2
- 101000654734 Homo sapiens Septin-4 Proteins 0.000 description 2
- 101000652747 Homo sapiens Target of rapamycin complex 2 subunit MAPKAP1 Proteins 0.000 description 2
- 101000648491 Homo sapiens Transportin-1 Proteins 0.000 description 2
- 102100032817 Integrin alpha-5 Human genes 0.000 description 2
- 108010008212 Integrin alpha4beta1 Proteins 0.000 description 2
- 108010041014 Integrin alpha5 Proteins 0.000 description 2
- 108010042918 Integrin alpha5beta1 Proteins 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- 101001027130 Mus musculus Fibronectin Proteins 0.000 description 2
- 101001043827 Mus musculus Interleukin-2 Proteins 0.000 description 2
- 102000002356 Nectin Human genes 0.000 description 2
- 108060005251 Nectin Proteins 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 101710176384 Peptide 1 Proteins 0.000 description 2
- 102100040969 Regulatory-associated protein of mTOR Human genes 0.000 description 2
- 102100032743 Septin-4 Human genes 0.000 description 2
- 108020004459 Small interfering RNA Proteins 0.000 description 2
- 210000000683 abdominal cavity Anatomy 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 230000000890 antigenic effect Effects 0.000 description 2
- 230000001640 apoptogenic effect Effects 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- 210000000601 blood cell Anatomy 0.000 description 2
- 239000007853 buffer solution Substances 0.000 description 2
- 210000004899 c-terminal region Anatomy 0.000 description 2
- 239000006143 cell culture medium Substances 0.000 description 2
- 238000009172 cell transfer therapy Methods 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 230000003399 chemotactic effect Effects 0.000 description 2
- 210000000349 chromosome Anatomy 0.000 description 2
- 230000000139 costimulatory effect Effects 0.000 description 2
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 210000004700 fetal blood Anatomy 0.000 description 2
- 238000010353 genetic engineering Methods 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 208000032839 leukemia Diseases 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 244000005700 microbiome Species 0.000 description 2
- 210000005087 mononuclear cell Anatomy 0.000 description 2
- 229940023041 peptide vaccine Drugs 0.000 description 2
- 210000005259 peripheral blood Anatomy 0.000 description 2
- 239000011886 peripheral blood Substances 0.000 description 2
- 230000002062 proliferating effect Effects 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 238000001959 radiotherapy Methods 0.000 description 2
- 239000012429 reaction media Substances 0.000 description 2
- 239000004055 small Interfering RNA Substances 0.000 description 2
- 229940054269 sodium pyruvate Drugs 0.000 description 2
- 230000003393 splenic effect Effects 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 238000002054 transplantation Methods 0.000 description 2
- 230000005740 tumor formation Effects 0.000 description 2
- 229960005486 vaccine Drugs 0.000 description 2
- 230000004580 weight loss Effects 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- DQJCDTNMLBYVAY-ZXXIYAEKSA-N (2S,5R,10R,13R)-16-{[(2R,3S,4R,5R)-3-{[(2S,3R,4R,5S,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy}-5-(ethylamino)-6-hydroxy-2-(hydroxymethyl)oxan-4-yl]oxy}-5-(4-aminobutyl)-10-carbamoyl-2,13-dimethyl-4,7,12,15-tetraoxo-3,6,11,14-tetraazaheptadecan-1-oic acid Chemical compound NCCCC[C@H](C(=O)N[C@@H](C)C(O)=O)NC(=O)CC[C@H](C(N)=O)NC(=O)[C@@H](C)NC(=O)C(C)O[C@@H]1[C@@H](NCC)C(O)O[C@H](CO)[C@H]1O[C@H]1[C@H](NC(C)=O)[C@@H](O)[C@H](O)[C@@H](CO)O1 DQJCDTNMLBYVAY-ZXXIYAEKSA-N 0.000 description 1
- NNRFRJQMBSBXGO-CIUDSAMLSA-N (3s)-3-[[2-[[(2s)-2-amino-5-(diaminomethylideneamino)pentanoyl]amino]acetyl]amino]-4-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-oxobutanoic acid Chemical group NC(N)=NCCC[C@H](N)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CO)C(O)=O NNRFRJQMBSBXGO-CIUDSAMLSA-N 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- 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 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
- 101710082414 50S ribosomal protein L12, chloroplastic Proteins 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 108090000672 Annexin A5 Proteins 0.000 description 1
- 102000004121 Annexin A5 Human genes 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000255789 Bombyx mori Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 108700031361 Brachyury Proteins 0.000 description 1
- 229910001369 Brass Inorganic materials 0.000 description 1
- 102100036842 C-C motif chemokine 19 Human genes 0.000 description 1
- 102100032367 C-C motif chemokine 5 Human genes 0.000 description 1
- 108010029697 CD40 Ligand Proteins 0.000 description 1
- 102100032937 CD40 ligand Human genes 0.000 description 1
- 210000001239 CD8-positive, alpha-beta cytotoxic T lymphocyte Anatomy 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 102000053642 Catalytic RNA Human genes 0.000 description 1
- 108090000994 Catalytic RNA Proteins 0.000 description 1
- 108010055166 Chemokine CCL5 Proteins 0.000 description 1
- 241001227713 Chiron Species 0.000 description 1
- 241000725101 Clea Species 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 235000019750 Crude protein Nutrition 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 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 1
- 241001131785 Escherichia coli HB101 Species 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 1
- 108700007698 Genetic Terminator Regions Proteins 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 102000002068 Glycopeptides Human genes 0.000 description 1
- 108010015899 Glycopeptides Proteins 0.000 description 1
- 102000025850 HLA-A2 Antigen Human genes 0.000 description 1
- 108010074032 HLA-A2 Antigen Proteins 0.000 description 1
- 206010018910 Haemolysis Diseases 0.000 description 1
- 241000162682 Heterogen Species 0.000 description 1
- 101000713106 Homo sapiens C-C motif chemokine 19 Proteins 0.000 description 1
- 101001002709 Homo sapiens Interleukin-4 Proteins 0.000 description 1
- 101000622137 Homo sapiens P-selectin Proteins 0.000 description 1
- 101000829980 Homo sapiens Ral guanine nucleotide dissociation stimulator Proteins 0.000 description 1
- 101000639987 Homo sapiens Stearoyl-CoA desaturase 5 Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- 108091006905 Human Serum Albumin Proteins 0.000 description 1
- 102000008100 Human Serum Albumin Human genes 0.000 description 1
- 108010047761 Interferon-alpha Proteins 0.000 description 1
- 102000006992 Interferon-alpha Human genes 0.000 description 1
- 108010066979 Interleukin-27 Proteins 0.000 description 1
- 102000000704 Interleukin-7 Human genes 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- 229930182816 L-glutamine Natural products 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 108700041567 MDR Genes Proteins 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 241000711408 Murine respirovirus Species 0.000 description 1
- 101001043808 Mus musculus Interleukin-7 Proteins 0.000 description 1
- 102100031789 Myeloid-derived growth factor Human genes 0.000 description 1
- 238000011887 Necropsy Methods 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 102100023472 P-selectin Human genes 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 102100023320 Ral guanine nucleotide dissociation stimulator Human genes 0.000 description 1
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 1
- DYAHQFWOVKZOOW-UHFFFAOYSA-N Sarin Chemical compound CC(C)OP(C)(F)=O DYAHQFWOVKZOOW-UHFFFAOYSA-N 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 241000580858 Simian-Human immunodeficiency virus Species 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 206010041925 Staphylococcal infections Diseases 0.000 description 1
- 102100033930 Stearoyl-CoA desaturase 5 Human genes 0.000 description 1
- 108010083312 T-Cell Antigen Receptor-CD3 Complex Proteins 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 108020004440 Thymidine kinase Proteins 0.000 description 1
- 241000244317 Tillandsia usneoides Species 0.000 description 1
- 108700009124 Transcription Initiation Site Proteins 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 108700025316 aldesleukin Proteins 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 238000005349 anion exchange Methods 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 230000030741 antigen processing and presentation Effects 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 229910052785 arsenic Inorganic materials 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 239000012503 blood component Substances 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 238000010322 bone marrow transplantation Methods 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 239000010951 brass Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 238000005341 cation exchange Methods 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000007969 cellular immunity Effects 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 239000013611 chromosomal DNA Substances 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 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 1
- 238000001647 drug administration Methods 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 238000002651 drug therapy Methods 0.000 description 1
- 210000003162 effector t lymphocyte Anatomy 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 230000005284 excitation Effects 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000012997 ficoll-paque Substances 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 238000002523 gelfiltration Methods 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 235000004554 glutamine Nutrition 0.000 description 1
- 239000001307 helium Substances 0.000 description 1
- 229910052734 helium Inorganic materials 0.000 description 1
- SWQJXJOGLNCZEY-UHFFFAOYSA-N helium atom Chemical compound [He] SWQJXJOGLNCZEY-UHFFFAOYSA-N 0.000 description 1
- 210000002443 helper t lymphocyte Anatomy 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 230000008588 hemolysis Effects 0.000 description 1
- 208000006454 hepatitis Diseases 0.000 description 1
- 231100000283 hepatitis Toxicity 0.000 description 1
- 230000006801 homologous recombination Effects 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 102000055229 human IL4 Human genes 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 238000010335 hydrothermal treatment Methods 0.000 description 1
- 230000003100 immobilizing effect Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 210000001821 langerhans cell Anatomy 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 230000006386 memory function Effects 0.000 description 1
- 208000015688 methicillin-resistant staphylococcus aureus infectious disease Diseases 0.000 description 1
- CSHFHJNMIMPJST-HOTGVXAUSA-N methyl (2s)-2-[[(2s)-2-[[2-[(2-aminoacetyl)amino]acetyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoate Chemical compound NCC(=O)NCC(=O)N[C@H](C(=O)N[C@@H](CC(C)C)C(=O)OC)CC1=CC=CC=C1 CSHFHJNMIMPJST-HOTGVXAUSA-N 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- OHDXDNUPVVYWOV-UHFFFAOYSA-N n-methyl-1-(2-naphthalen-1-ylsulfanylphenyl)methanamine Chemical compound CNCC1=CC=CC=C1SC1=CC=CC2=CC=CC=C12 OHDXDNUPVVYWOV-UHFFFAOYSA-N 0.000 description 1
- 210000000581 natural killer T-cell Anatomy 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 239000004417 polycarbonate Substances 0.000 description 1
- 229920000515 polycarbonate Polymers 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 229940087463 proleukin Drugs 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 108010056030 retronectin Proteins 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 108091092562 ribozyme Proteins 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 238000000527 sonication Methods 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 210000004988 splenocyte Anatomy 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 206010052366 systemic mycosis Diseases 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 201000008827 tuberculosis Diseases 0.000 description 1
- 230000036326 tumor accumulation Effects 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 238000005406 washing Methods 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
- 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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/462—Cellular immunotherapy characterized by the effect or the function of the cells
- A61K39/4621—Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/46434—Antigens related to induction of tolerance to non-self
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/46449—Melanoma antigens
- A61K39/464491—Melan-A/MART
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/464499—Undefined tumor antigens, e.g. tumor lysate or antigens targeted by cells isolated from tumor
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/10—Antimycotics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
- A61P31/16—Antivirals for RNA viruses for influenza or rhinoviruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/04—Immunostimulants
-
- 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
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- 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
- C12N5/10—Cells modified by introduction of foreign genetic material
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/50—Cell markers; Cell surface determinants
- C12N2501/515—CD3, T-cell receptor complex
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/50—Cell markers; Cell surface determinants
- C12N2501/58—Adhesion molecules, e.g. ICAM, VCAM, CD18 (ligand), CD11 (ligand), CD49 (ligand)
-
- 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
- C12N2533/00—Supports or coatings for cell culture, characterised by material
- C12N2533/50—Proteins
- C12N2533/52—Fibronectin; Laminin
-
- 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
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/10041—Use of virus, viral particle or viral elements as a vector
- C12N2740/10043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Definitions
- the present invention relates to a method for producing a T cell population useful in the medical field.
- B lymphocytes B cells
- T lymphocytes T cells
- T cells The majority of T cells are CD4 T cells having a CD (Cluster of Differentiation) 4 marker and CD8 T cells having a CD8 marker in the periphery.
- CD4T cells The majority of CD4T cells are called helper T cells (hereinafter referred to as T), which assists in antibody production and provides various types of relief.
- Thl type or Th2 type which have different types of site force-in produced by antigen stimulation.
- the majority of CD8 T cells are cytotoxic T cells that exhibit cytotoxic activity upon antigen stimulation [Tc: cytotoxic T lymphocytes, also known as killer T cells, hereinafter referred to as CTLs. ]
- immunotherapy has recently attracted attention as the fourth treatment method after surgery, chemotherapy, and radiation therapy in the pathology of cancer. Since immunotherapy uses the immunity inherent in humans, it is said that the physical burden on patients is light compared to other treatment methods.
- CTLs and peripheral blood lymphocytes, etc. induced outside the body, therapy for transferring lymphokine-active cells, NKT cells, ⁇ cells, etc. obtained by various methods of expansion, antigens in the body Known are cell-cell transfer therapy, peptide vaccine therapy, and Thl cell therapy, which are expected to induce specific CTLs, as well as immune gene therapy that introduces genes that can be expected to have various effects in these cells and transfers them into the body. Be beaten!
- Fibronectin exists in the blood of animals, on the surface of cultured cells, and in the extracellular matrix of tissues. It is a huge glycoprotein with a molecular weight of 250,000 and is known to have various functions. The domain structure is divided into seven parts (see Fig. 1 below), and the amino acid sequence contains three types of similar sequences. ing. Three types of similar sequences are called type I, type II, and type ⁇ ⁇ . Of these, type ⁇ is composed of 71-96 amino acid residues, and the concordance rate of these amino acid residues is 17- 40%.
- III-8, III-9 and III-10 The power of 14 type III sequences in fibronectin, of which 8th, 9th and 10th (hereinafter referred to as III-8, III-9 and III-10, respectively) are cell binding domains,
- the 12th, 13th, and 14th (hereinafter referred to as III-12, III13, and III-14, respectively) are contained in the heparin-binding domain.
- III-10 contains a very active activation antigen (VLA) -5 binding region, and this core sequence is RGDS.
- VLA very active activation antigen
- IIICS exists on the C-terminal side of the binding domain.
- III CS has a region called CS-1 that has binding activity to VLA-4, which has a capacity of 25 amino acids (for example, Non-Patent Documents 1 to 3).
- lymphokine-active cells derived from expanded CTLs and peripheral blood lymphocytes derived from outside the body by the action of IL-2 and anti-CD3 antibodies are transferred to V ⁇ .
- problems such as maintaining much cytotoxic activity when expanding antigen-specific CTLs induced outside the body, or how efficiently lymphocytes can be expanded outside the body, fibronectin and its fragments can be used.
- the effects of use have already been studied by the present inventors (for example, Patent Documents 1 to 3).
- T lymphocytes used for immunotherapy have been administered naive T cells or central memory T cells that are more indeterminate than effector T cells that have already been terminally separated.
- naive T cells or central memory T cells that are more indeterminate than effector T cells that have already been terminally separated.
- rod-like cell transfer therapy and peptide vaccine therapy that are expected to induce antigen-specific CTL in the body, for example, naive T that can be induced by CTL in patients with advanced cancer Because there are few cells, sufficient effects are often not expected.
- Non-Patent Literature l Deane F. Momer, published in 1988, FIBRONECTIN, ACA DEMIC PRESS INC., Pl-8
- Non-Patent Document 2 Kimizuka F. and 8 others, Biochem., 1991, Vol. 110, No. 2, p284-291
- Non-Patent Document 3 Hanenberg H. and 5 others, Human Gene Therapy, 1997, Vo 1. 8, No. 18, p2193-2206
- Non-Patent Document 4 Gattinoni L. and 9 others, J. Clin. Invest. 2005, Vol. 115, No. 6, P1616-1626
- Non-Patent Document 5 Benigni F and 10 others, Immunol. 2005, Vol. 175, No. 2, P739-748
- Patent Document 1 International Publication No. 03Z016511 Pamphlet
- Patent Document 2 Pamphlet of International Publication No. 03Z080817
- Patent Document 3 International Publication No. 2005Z019450 Pamphlet
- An object of the present invention is to provide a method for producing a T cell population effective for administration to a living body.
- a first invention of the present invention expresses CD45RA, comprising a step of culturing a cell population containing T cells in the presence of fibronectin, a fragment thereof or a mixture thereof, And a method for producing a T cell population that expresses at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28.
- examples of the total culture days including the culturing step include 4 to 14 days.
- the culture in the presence of fibronectin, a fragment thereof or a mixture thereof is exemplified at least at the beginning of the culture, and the culture is preferably performed for at least one day.
- the step of culturing in the presence of hive mouth nectin, a fragment thereof or a mixture thereof is carried out in the presence of CD3 ligand.
- the CD3 ligand include anti-CD3 antibodies.
- the fibronectin fragment comprises at least one amino acid sequence represented by SEQ ID NOs: 1 to 8 in the sequence listing. It is a polypeptide (m) or a polypeptide comprising at least one amino acid sequence in which one or more amino acids are substituted, deleted, inserted or added in any one of the above amino acid sequences.
- a polypeptide (n) having a function equivalent to that of the polypeptide (m) is exemplified.
- fibronectin fragments include polypeptides comprising any of the amino acid sequences represented by SEQ ID NOs: 1 to 3 and 5 to 8 in the sequence listing.
- a production method including the step of separating cells expressing at least one selected from the group consisting of CD45RA, CD62L, CCR7, CD27 and CD28.
- a production method further including the step of introducing a foreign gene into a cell population is exemplified.
- a retrovirus vector, an adenovirus vector, an adeno-associated virus vector, a lentivirus vector, or a simian virus vector can be used for introducing a foreign gene.
- At least one selected from the group consisting of CD62L, CCR7, CD27 and CD28 which is obtained by the method of the first aspect of the present invention and expresses CD45RA. It relates to T cell populations that are expressed.
- the third invention of the present invention expresses CD45RA obtained by the first invention of the present invention, and expresses at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28.
- the present invention relates to a medicine containing a T cell population as an active ingredient.
- the subject is selected from the group consisting of CD62L, CCR7, CD27 and CD28 that expresses an effective amount of CD45RA obtained by the method of the first invention of the present invention.
- the present invention also relates to a method for treating or preventing a disease comprising the step of administering a T cell population expressing at least one of the above.
- a fifth invention of the present invention is selected from the group consisting of CD62L, CCR7, CD27, and CD28, which is obtained by the method of the first invention of the present invention for the manufacture of a medicament, and which expresses CD45RA.
- the sixth invention of the present invention comprises at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28, which is obtained by the method of the first invention of the present invention and expresses CD45RA.
- the present invention relates to a method for producing a T cell population.
- a seventh invention of the present invention relates to a T cell population obtained by the method of the sixth invention of the present invention.
- the eighth invention of the present invention relates to a medicament comprising, as an active ingredient, a T cell population obtained by the method of the sixth invention of the present invention.
- the ninth invention of the present invention relates to a method for treating or preventing a disease comprising the step of administering to a subject an effective amount of a T cell population obtained by the method of the sixth invention of the present invention.
- the tenth invention of the present invention relates to the use of the T cell population obtained by the method of the sixth invention of the present invention for the manufacture of a medicament.
- the eleventh invention of the present invention is a small molecule selected from the group consisting of (a) CD45RA obtained by the method of the first invention of the present invention and consisting of CD62L, CCR7, CD27 and CD28.
- a pharmaceutical comprising a preparation containing at least one selected stimulator as an active ingredient, wherein the preparation comprises two separate preparations to be administered simultaneously or separately.
- a twelfth aspect of the present invention relates to a method for treating a disease, which comprises the following steps (a) and (b).
- the production method of the present invention provides a T cell population that expresses CD45RA and expresses at least one selected from the group consisting of CD62L, CCR7, CD27, and CD28.
- the cell population obtained by the production method is used to treat diseases by cell therapy in which the ratio of T cells expressing CD45RA and expressing at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28 is high. Very useful.
- the present invention includes a step of culturing in the presence of fibronectin, a fragment thereof or a mixture thereof (hereinafter sometimes referred to as an active ingredient of the present invention), thereby expressing CD45RA and CD62L
- the present inventors have found that a cell population containing a high proportion of T cells expressing at least one selected from the group consisting of CCR7, CD27 and CD28 can be obtained, and has been completed.
- the T cell population expressing CD45RA and expressing at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28 expresses CD45RA
- the high ratio here means that CD45 in the T cell population obtained by culturing in the presence of the active ingredient of the present invention when culturing was carried out under the same conditions except for the presence or absence of the active ingredient of the present invention.
- the ratio of T cells that express RA and express at least one selected from the group consisting of CD62L, CCR7, CD27, and CD28 is higher than that in the absence of the active ingredient of the present invention.
- the T cell population is preferably 5% or more, more preferably 10% or more higher than that in the absence of the active ingredient of the present invention.
- the proportion of T cells that express CD45RA and express at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28 in the resulting cell population depends on various environmental factors such as peripheral blood mononuclear Since it varies depending on individual differences and physical condition of humans supplying cells used for the production of T cells such as spheres (PBMC), it is not possible to specify the above ⁇ high ratio '' by numerical values. Is possible.
- the T cell population obtained by the production method of the present invention means a population containing T cells, and cells other than T cells, such as NK cells. Other lymphocytes such as vesicles and blood cell components other than lymphocyte
- the fibronectin and fragments thereof described herein may be either naturally occurring or artificially synthesized. Fibronectin and fragments thereof are described, for example, in Ruoslahti E. et al. (Ruoslahti E., et al., J. Biol. Chem. -7281 (1981)], it can be produced in substantially pure form from naturally occurring substances. As used herein, the substantially pure fib mouthctin or fibronectin fragment described herein essentially contains other proteins that are naturally present with fibronectin. Means no. The above fibronectin and fragments thereof can be used in the present invention singly or as a mixture of plural kinds.
- fibronectin is known to have many splicing variants, any variant may be used as the fibronectin as long as it exhibits the desired effect of the present invention. be able to.
- plasma-derived hive mouth nectin lacks the region called ED-B that exists upstream of the cell-binding domain, and the region called ED-A that exists between the cell-binding domain and the heparin-binding domain. It is known that! /, However, such plasma-derived fibronectin can also be used in the present invention.
- Fibronectin fragments that can be used in the present invention, as well as useful information regarding the preparation of the fragments, are described in Kimika F. et al. [13 ⁇ 41 ⁇ (1111 ⁇ F., et al., Journal 'Ob' Biochemistry ( J. Biochem.), 110, 284-291 (1991)], Cornbright AR et al. [13 ⁇ 41: 111 ⁇ 111 AR, et al., EMBO Journal (EMBO J.), IV, 7 , 1755-1759 (1985)], and Sekiguchi K. 3 ⁇ 4 [Sekiguchi K., et al., Biochemistry, No. 25, No. 17, 4936-494 1 (1986)], etc.
- Genbank Accession No. NM 0 02026, NP 1 002017 for the nucleic acid sequence encoding fibronectin or the amino acid sequence of fibronectin.
- fibronectin fragments include, for example, III-8 (amino acid sequence represented by SEQ ID NO: 1 in the sequence listing), ⁇ -9 (amino acid sequence represented by SEQ ID NO: 2 in the sequence listing) ), III-10 (amino acid sequence represented by SEQ ID NO: 3 in the sequence listing), ⁇ —11 (amino acid sequence represented by SEQ ID NO: 4 in the sequence listing), III 12 (represented by SEQ ID NO: 5 in the sequence listing) Amino acid sequence), ⁇ —13 (amino acid sequence represented by SEQ ID NO: 6 in the sequence listing), ⁇ —14 (amino acid sequence represented by SEQ ID NO: 7 in the sequence listing), and CS—1 (sequence listing)
- a polypeptide (m) comprising at least one amino acid sequence constituting any region of the amino acid sequence represented by SEQ ID NO: 8 (see Fig.
- amino acid sequence comprising at least one comprising at polypeptides, Poribe peptide having the same function as the polypeptide (m) (n) is illustrated.
- the number of amino acids is 20 to 1000 force, more preferably 100 to 800 force.
- the term “multiple” is a concept including several pieces, 2 to 12 pieces are preferred, 2 to 10 pieces are more preferred, and 2 to 8 pieces are more preferred. It is the same.
- the fragment those having cell adhesion activity and Z or hen binding activity can be preferably used.
- the cell adhesion activity can be examined by assessing the binding between the fragment used in the present invention (its cell binding domain) and the cell using a known method.
- such methods include the method of Williams D. A. et al. [Williams D. A., et al., Nature, 352, 438-441 (199 1)].
- This method is a method for measuring the binding of cells to a fragment immobilized on a culture plate.
- the heparin binding activity can be examined by assessing the binding of the fragment used in the present invention (its heparin binding domain) to heparin using a known method.
- fibronectin fragments include C274 (amino acid sequence represented by SEQ ID NO: 9 in the sequence listing), H-271 (amino acid sequence represented by SEQ ID NO: 10 in the sequence listing), H-2 96 (amino acid sequence represented by SEQ ID NO: 11 in the sequence listing), CH-271 (amino acid sequence represented by SEQ ID NO: 12 in the sequence listing), CH-296 (amino acid sequence represented by SEQ ID NO: 13 in the sequence listing) Column), C—CS1 (amino acid sequence represented by SEQ ID NO: 14 in the Sequence Listing), and CH—296N a (amino acid sequence represented by SEQ ID NO: 15 in the Sequence Listing). Chido is exemplified.
- Each fragment of CH-271, CH-296, CH-296Na, C-274, and C-CS1 is a polypeptide having a cell-binding domain having an activity of binding to VLA-5.
- C-CS1, H-296, CH-296, and CH-296Na are polypeptides having CS-1 having an activity of binding to VLA-4.
- H-271, H-296, CH-271, CH-296 and CH-296Na are polypeptides having a heparin-binding domain.
- CH-296Na is a polypeptide containing from cell binding domain to C S 1 in fibronectin derived from plasma.
- a fragment in which each of the above domains is modified can also be used.
- the heparin-binding domain of fibronectin is composed of three type III sequences ( ⁇ -12, III-13, 1 11-14).
- a fragment containing a heterogen binding domain from which one or two of the type III sequences have been deleted can also be used in the present invention.
- a cell binding site of fibronectin VLA-5 binding region, Prol239 to Serl515) and one type III sequence CHV-89 (amino acid sequence represented by SEQ ID NO: 16 in the sequence listing), CHV—90 (amino acid sequence represented by SEQ ID NO: 17 in the Sequence Listing), CHV—92 (amino acid sequence represented by SEQ ID NO: 18 in the Sequence Listing), or CHV that is a fragment obtained by combining two ⁇ -type sequences — 179 (amino acid sequence represented by SEQ ID NO: 19 in the Sequence Listing), CHV—181 (amino acid sequence represented by SEQ ID NO: 20 in the Sequence Listing).
- CHV-89, CHV-90, and CHV-92 contain III-13, 111-14, and III-12, respectively, CHV-179 contains III-13 and III-14, and CHV-181 contains III-12. And III—1 3 respectively.
- a fragment in which an amino acid is further added to each of the above fragments can also be used in the present invention.
- the fragment can be produced, for example, by adding a desired amino acid to each of the above fragments.
- H—275—Cys SEQ ID NO: 21 Is a fragment having a heparin binding domain of fibronectin and a cysteine residue at the C-terminal.
- the fragment used in the present invention has a function equivalent to that of the fragment containing at least a part of the amino acid sequence of the natural fibronectin exemplified above as long as the desired effect of the present invention is obtained. It may be composed of a polypeptide having an amino acid sequence having substitution, deletion, insertion or addition of one or more amino acids in the amino acid sequence of the polypeptide constituting the fragment.
- amino acid substitution and the like are preferably such that the physical and physical properties of the polypeptide can be changed within a range in which the function of the original polypeptide can be maintained.
- substitution etc. of amino acids the nature (e.g., hydrophobic, hydrophilic, electrostatic load, P K, etc.) peculiar to a polypeptide that substantially conservative range that does not change is preferably a.
- amino acid substitutions are: 1. Glycine, alanine; 2. Norin, isoleucine, leucine; 3. Aspartic acid, glutamic acid, asparagine, glutamine; 4. Serine, threonine; 5.
- Lysine arginine
- amino acid deletions, additions, and insertions are those of amino acids with properties similar to those around the target site in the polypeptide. Deletions, additions and insertions within the range that does not substantially change the properties are preferred.
- the fragment used in the present invention is obtained by genetic engineering, for example, when producing Escherichia coli or the like as a host, methionine at the heel end is affected by the influence of methionine beptidase derived from Escherichia coli.
- Such polypeptides may also be used in the present invention although they may be deleted. That is, polypeptides lacking the methionine at the terminus of the polypeptides described in SEQ ID NOs: 15 and 21 in the sequence listing can also be suitably used in the present invention.
- the amino acid substitution or the like may be naturally occurring due to interspecies or individual differences, or may be artificially induced. Artificial induction is not particularly limited as long as it can be performed by a known method.
- one or more bases are substituted in a nucleic acid encoding the above-mentioned region derived from natural fibronectin or a predetermined fragment by a known method. Make a given nucleic acid, deleted, added or inserted, and use it to A polypeptide comprising an amino acid sequence having a function equivalent to that of the above fibronectin or a predetermined fragment and having a substitution in the amino acid sequence of the polypeptide constituting the fragment or the like can be produced.
- “having an equivalent function” refers to a group that expresses CD45RA in a T cell population obtained using a polypeptide and is composed of CD62L, CCR7, CD27, and CD28.
- the ratio of T cells expressing at least one of the more selected is higher than the T cell population obtained in the absence of the control polypeptide.
- the above action can be appropriately confirmed according to the methods described in Examples 1, 2, and 6 described later.
- those having cell adhesion activity and z or heparin binding activity are preferred, and those having a CS-1 domain are also preferred.
- Cell adhesion activity and amino acid binding activity can be evaluated according to the above-described method for measuring activity.
- fragment comprising a polypeptide having an amino acid substitution or the like
- a fragment in which one or more amino acids are inserted as a linker between two different domains can also be used in the present invention.
- fibronectin is also a polypeptide having an amino acid sequence having one or more amino acid substitutions, deletions, insertions or additions in the amino acid sequence of the polypeptide, similar to the above fragment, Ratio of T cells expressing CD45RA in a T cell population obtained using the polypeptide and expressing at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28 Polypeptides that are higher than the T cell population obtained in the absence of tides can be used in the present invention.
- the fibronectin or fragment thereof used in the present invention is equivalent to the natural fibronectin exemplified above and a fragment containing at least a part of its amino acid sequence as long as the desired effect of the present invention is obtained.
- a polypeptide having 50% or more homology with the amino acid sequence of the polypeptide constituting the fibronectin or a fragment thereof having a unique function preferably a polypeptide having 70% or more homology, more preferably 90%
- a polypeptide having the above homology, more preferably 95% or less Peptides having the above homology can be used.
- DNASIS Pro Ver. 2.6 manufactured by Takara Bio Inc.
- fibronectin fragments most preferably used include ⁇ -8 (amino acid sequence represented by SEQ ID NO: 1 in the sequence listing) and III 9 (sequence listing in the sequence listing) in the amino acid sequence.
- Amino acid sequence represented by number 2) ⁇ —10 (amino acid sequence represented by SEQ ID NO: 3 in the sequence listing), ⁇ —12 (amino acid sequence represented by SEQ ID NO: 5 in the sequence listing), ⁇ —13 ( Amino acid sequence represented by SEQ ID NO: 6 in the sequence listing), ⁇ —14 (amino acid sequence represented by SEQ ID NO: 7 in the sequence listing) and CS—1 (amino acid sequence represented by SEQ ID NO: 8 in the sequence listing).
- Fragments containing all of them that is, polypeptides containing a heparin-binding domain, a cell-binding domain, and CS-1, and more preferably having the same function as CH-296 or CH-296 described above
- Polypeptides containing a heparin-binding domain, a cell-binding domain, and CS-1 make up poly Examples thereof include a polypeptide having an amino acid sequence having substitution, deletion, insertion or addition of one or more amino acids in the amino acid sequence of the peptide.
- the fibronectin polypeptide suitably used in the present invention has H-296, CH-271, H-271, C-CS1, or a function equivalent to these as shown in Example 18.
- the fibronectin fragment described in the present specification is produced as a recombinant fibronectin fragment from a gene recombinant based on, for example, the description in US Pat. No. 5,198,423.
- the C-274 (SEQ ID NO: 9) fragment can be obtained by the method described in US Pat. No. 5,102,988. Furthermore, the C—CS1 (SEQ ID NO: 14) fragment can be obtained by the method described in Japanese Patent No. 3104178.
- the above fragments of CHV-89 (SEQ ID NO: 16), CHV-90 (SEQ ID NO: 17), and CHV-179 (SEQ ID NO: 19) are disclosed in Japanese Patent No. 2729712. It can be obtained by the method described in the specification.
- the CHV-181 (SEQ ID NO: 20) fragment can be obtained according to the method described in WO 97Z18318 pamphlet.
- For the CHV-92 (SEQ ID NO: 18) fragment refer to Japanese Patent No. 2729712 and the pamphlet of International Publication No. 97Z18318. Based on the plasmids described in those documents, the plasmid was routinely constructed. The plasmid can be used for genetic engineering.
- FERM BP-2264 Escherichia coli carrying a plasmid encoding H-271; deposit date 19 Jan. 30, 1989
- FERM BP-2800 Escherichia coli carrying a plasmid encoding CH-296; deposit date 12 May 1989
- FERM BP-2799 E. coli carrying a plasmid encoding CH-271; deposit date 12 May 1989
- FERM BP—7420 E. coli harboring a plasmid encoding H—296; deposit date 19 May 12 12
- FERM BP— 1915 Escherichia coli carrying a plasmid encoding C— 274; date of deposit 19 June 1888
- FERM BP— 5723 Escherichia coli carrying a plasmid encoding C—CS1; deposit date 1 March 5th, 990
- FERM BP—10073 (plasmid encoding CH—296Na; date of deposit, July 2, 2004)
- FERM P—12182 E. coli carrying a plasmid encoding CHV—89; deposit date 8 April 1991
- E. coli harboring a plasmid encoding FERM P-12183 (CHV-179; date of deposit 1991 April 8th).
- fibronectin is a large glycoprotein, it is not always easy to prepare and use a protein of natural origin from an industrial and pharmaceutical standpoint.
- fibronectin is a multifunctional protein, it may be caused by inconvenience due to a region different from the region that is effective for the method of the present invention depending on the situation of its use. From these, in the present invention, from the viewpoint of availability, ease of handling, and safety, it is preferable to use a fibronectin fragment, more preferably a recombinant fibronectin fragment obtained as described above. It is preferable to do. Also, from the viewpoint of realizing a high expansion culture rate, it is preferable to use the aforementioned fibronectin fragment.
- the molecular weight of the fibronectin fragment used in the present invention is not particularly limited, but is preferably 1 to 200 kD, more preferably 5 to 190 kD, and further preferably 10 to 180 kD.
- the molecular weight can be determined, for example, by SDS-polyacrylamide gel electrophoresis.
- amino acid sequence of the polypeptide constituting the fibronectin fragment of the present invention the amino acid sequence portion other than the amino acid sequence of the polypeptide constituting the naturally derived fibronectin fragment is the desired effect of the present invention. It is optional as long as it does not inhibit the expression of, and is not particularly limited.
- the present invention expresses CD45RA and T cells expressing at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28, preferably expressing CD45RA and from the group consisting of CD62L and CCR7.
- This is a method for producing a cell population containing a high ratio of T cells expressing at least one selected.
- the method of the present invention comprises the step of culturing a cell population containing T cells in the presence of the aforementioned fibronectin, a fragment thereof or a mixture thereof.
- CD45RA, CD62L, CCR7, CD27, and CD28 are all cell surface antigen markers for lymphocytes, and are known to be expressed in undifferentiated cells such as naive T cells. That is, it is contained in a high ratio in the cell population obtained by the production method of the present invention.
- T cells that express CD45RA and express at least one selected from the group consisting of CD62L, CCR7, CD27, and CD28 can be differentiated into memory T cells according to the phenotype of the cell surface antigen marker. Can be classified as undifferentiated cells, that is, na ⁇ ve T-like cells.
- naive T cells are in vivo survival when administered to a living body, cell proliferation effect, tumor accumulation effect, tumor-specific effector cell production rate It is described that it is useful in the field of cell therapy.
- the T cell population obtained by the production method of the present invention produces a large amount of IL-2 by stimulation with anti-CD3 antibody or anti-CD28 antibody as shown in Example 3 described later. Become activated T cells.
- the T cell population obtained by the production method of the present invention reacts with chemokine CCL21 and exhibits chemotaxis, so that it has the ability to migrate to lymph nodes. It also has.
- CTL having antigen-specific cytotoxic activity is induced by applying antigen stimulation to the T cell population.
- the T cell population is in the presence or absence of a small amount of IL-2 compared to a T cell population produced in the absence of fibronectin, a fragment thereof or a mixture thereof. Since survival in the presence is high, survival in vivo is expected to be high.
- Example 11 described below when the T cell population obtained by the production method of the present invention is administered to NODZscid mice, the T cell population produced in the absence of fibronectin, a fragment thereof or a mixture thereof is used. Compared to administration, T cells have a higher engraftment rate in the spleen and a higher survival rate.
- the administered T cell population also induces a GVHD response.
- the T cell population obtained by the production method of the present invention has not only a phenotype of cell surface antigen markers, but also functions suitable for use in the field of cell medicine possessed by naive T cells. RU
- T cell population obtained by the process of separating the cell population that expresses the phenotype is compared with naive T cells obtained from PBMC and T cells showing the same phenotype obtained in the absence of the active ingredient of the present invention. Therefore, the cytotoxic activity of induced CTL is high and the specific antigen recognition ability is also high.
- the T cell population obtained by such a separation procedure has a significant increase in cytotoxic activity when separated into CTL compared to normal na ⁇ ve T cells. It is a cell population comprising a novel naive T-like cell with more effective characteristics different from naive T cells.
- the high therapeutic effect of the T cell population obtained by the production method of the present invention as described above is expected in the same way for both CD8 + and CD4 + naive T-like cells. Furthermore, as described in the above-mentioned Patent Document 2, it is possible to achieve a very high proliferation rate by expanding T cells in the presence of fibronectin, a fragment thereof or a mixture thereof.
- the T cell population obtained by the method of the invention is very suitable for use in the field of cell medicine.
- Examples of the cell population containing T cells used in the production method of the present invention include PBMC, naive T cells, memory T cells, hematopoietic stem cells, cord blood mononuclear cells and the like.
- any blood cell can be used in the present invention.
- These cells may be any of those obtained by collecting vitality, or those obtained through in vitro culture, for example, those obtained by directly or cryopreserving the T cell population obtained by the method of the present invention. it can.
- the cell force used in the production of the above-mentioned T cell population obtained by vital force a cell population obtained through various separation operations, for example, a cell such as PBMC is obtained by separating into CD8 + or CD4 + cells.
- any of the specified cell populations can also be used.
- a material containing the above cells for example, blood such as peripheral blood and umbilical cord blood, a material obtained by removing components such as red blood cells and plasma from blood, bone marrow fluid, etc. is used. can do.
- the total number of culture days is 4 to 14 days. That is, when the total number of culture days is 4 to 14 days, T cells expressing CD45RA and expressing at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28 in the obtained T cell population For use in the field of cellular medicine Very suitable. If the total number of culture days is less than 4 days, it will not be possible to obtain a satisfactory number of cells for use in general immunotherapy. In the present invention, the total number of culture days is preferably 5 to 14 days, more preferably 7 to 14 days.
- the culture is performed in the presence of the active ingredient of the present invention at least at an initial stage during the entire culture period. It is preferable that the culture is carried out in the presence of the active ingredient of the present invention at least at the start of the culture.
- the culture in the presence of the active ingredient of the present invention may be the entire period during the culture period or may be an arbitrary partial period. That is, the present invention is included in the present invention as long as the process is included in a part of the T cell production process.
- the culture in the presence of the active ingredient of the present invention is preferably carried out at least at least 1 day, more preferably at least 3 days, even more preferably at least 4 days.
- the concentration of fibronectin, a fragment thereof, or a mixture thereof in the culture is not particularly limited.
- 0.001-500 ⁇ g / mL, particularly 0.01-500 ⁇ m. gZmL is preferred.
- fibronectin a fragment thereof, or a mixture thereof is used from the viewpoint of effectively stimulating the TCR-CD3 complex of T cells and proliferating the cells. It is preferable to perform the culture in the presence of CD3 ligand! / ,.
- the CD3 ligand is not particularly limited as long as it is a substance having binding activity to CD3.
- an anti-CD3 antibody is exemplified, and an anti-CD3 monoclonal antibody is particularly preferably exemplified.
- OKT3 is exemplified.
- the concentration of CD3 ligand in the medium is not particularly limited.
- an anti-CD3 monoclonal antibody for example, 0.0.OOl ⁇ lOO.ug / mL, particularly 0.01 to: LOO / z gZmL force is suitable. is there.
- a costimulation can be introduced by adding other costimulation factors such as CD28 ligand.
- costimulation factors such as CD28 ligand.
- CD28 ligand for example, anti-CD28 antibody, CD80, B7-1, B7-2 and the like are exemplified.
- the medium used in the method for producing a T cell population of the present invention is not particularly limited, and a known medium prepared by mixing components necessary for expansion culture of T cells can be used.
- a commercially available medium can be appropriately selected and used.
- These media are In addition to the original constituents, it may contain cytosines, suitable proteins, and other components.
- the cytodynamic ins include IL-2, IL-7, IL-12, IFN-y and the like, and a medium containing IL2 is preferably used.
- the concentration of IL 2 in the medium is not particularly limited, and is preferably 0.01 to 1 ⁇ 10 5 U / mL, more preferably 0.1 to 1 ⁇ 10 4 UZmL, for example.
- suitable proteins include anti-IL-4 antibodies.
- lymphocyte stimulating factors such as lectins can be added.
- the concentration of the component in the medium is not particularly limited as long as a desired effect is obtained.
- serum or plasma can be added to the medium.
- the amount added to these media is not particularly limited, but is 0 to 20% by volume, and the amount of serum or plasma used can be changed according to the culture stage.
- the serum or plasma concentration can be decreased in stages.
- the origin of serum or plasma may be either self (meaning that the origin is the same as the cell being cultured) or non-self (meaning that the origin is different from the cell being cultured), Preferably, self-derived ones can be used from the viewpoint of safety.
- the T cell population of the present invention is usually produced in a medium containing a predetermined component in the presence of the active ingredient of the present invention.
- the number of cells at the start of the culture used in the present invention is not particularly limited. For example, it is preferably lcellZmL to l ⁇ 10 8 cells / mL, more preferably lcell / mL to 5 ⁇ 10 7. cells / mL, more preferably it is exemplified lcell / mL ⁇ 2 X 10 7 Ce llsZmL .
- the conditions used for normal cell culture can be used. For example, culture under conditions of 37 ° C, 5% CO, etc.
- the cell culture solution can be diluted at an appropriate time interval by adding a fresh medium.
- the power for exchanging the medium or the cell culture equipment can be exchanged.
- the cell culture equipment used in the method for producing a T cell population of the present invention is not particularly limited.
- a petri dish, a flask, a nog, a large culture tank, a bioreactor, etc. may be used. it can.
- NOGG CO gas permeable bag for cell culture
- a large culture tank can be used. Cultivation should be performed in either open or closed systems. It is preferable to culture in a closed system from the viewpoint of the safety of the obtained T cells.
- the active ingredient of the present invention CD3 ligand, other costimulatory factors, suitable proteins contained in the above-mentioned medium, cytosines, and other ingredients can be dissolved and coexisted in the medium.
- a suitable solid phase eg cell culture equipment such as petri dishes, flasks, bags, etc. (including both open and closed systems), or cell culture carriers such as beads, membranes, slide glass, etc. You may use it.
- the material of the solid phase is not particularly limited as long as it can be used for cell culture.
- the ratio is the same as the desired concentration when the component is dissolved in the medium.
- the amount of the component to be immobilized is not particularly limited as long as a desired effect is obtained.
- the carrier is used by immersing it in a culture medium in a cell culture equipment during cell culture.
- the component is immobilized on the carrier, when the carrier is placed in the medium, it is placed in the equipment so that the ratio is the same as the desired concentration when the component is dissolved in the medium.
- the amount of immobilization of the component is not particularly limited as long as a desired effect is obtained.
- the method for immobilizing the active ingredient of the present invention, CD3 ligand, and other costimulatory factors on the solid phase is not particularly limited.
- these substances may be immobilized in a suitable buffer solution in the solid phase. It is possible to fix it by insulting it.
- the immobilization of the fibronectin fragment to the solid phase can also be carried out by the methods described in WO97Z18318 pamphlet and WO00Z09168 pamphlet.
- T cell population is obtained by the method of the present invention, and then the T cell population and the solid phase are separated.
- the active ingredient and the T cell population are separated from each other and prevent the active ingredient and the like from being mixed into the T cell population.
- the production method of the present invention comprises a T cell obtained by culturing in the presence of the active ingredient of the present invention.
- the cell population may further comprise a step of separating a T cell population expressing at least one selected from the group consisting of CD45RA, CD62L, CCR7, CD27 and CD28. That is, as described above, the T cell population obtained by culturing in the presence of the active ingredient of the present invention expresses CD45RA at a high ratio and at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28. Separation of cells that express at least one surface antigen marker selected from the group consisting of CD45RA, CD62L, CCR7, CD27 and CD28.
- the T cells to be isolated are not particularly limited, but examples include cells expressing CD45RA, preferably expressing CD45RA and selected from the group consisting of CD62L, CCR7, CD27 and CD28. Examples include cells expressing at least one, more preferably cells expressing CD45RA and CD62L, and cells expressing CD45RA and CCR7.
- the separation operation is not particularly limited. For example, separation can be performed by a known method using a cell sorter, magnetic beads, a column, or the like. For example, separation of cells expressing CD45RA and CCR7 can be performed as described in Example 3- (3) below.
- the T cells produced by the method of the present invention can be cloned and maintained as stable T cells.
- a new ⁇ cell population can be obtained.
- an antigen-specific CTL can be produced by applying antigen stimulation or the like by a known method, for example, the same method as in Examples 5 to 8 described later.
- the T cell population obtained by the method of the present invention expresses at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28, which expresses undifferentiated T cells, CD45RA as described above.
- T cells having cytotoxic activity are also included in the T cell population.
- the cytotoxic activity of the T cell population can be evaluated by a known in vitro test, the T cell population obtained by the present invention is an undifferentiated naive T-like as described above. Since the cells are contained in a high ratio, the ⁇ cell population obtained by the production method of the present invention does not necessarily exhibit high cytotoxic activity in such an evaluation system.
- the disease to which the T cell population produced by the method of the present invention is administered is not particularly limited.
- cancer leukemia, malignant tumor, hepatitis, viruses such as influenza and HIV, bacteria, fungi
- infectious diseases caused by the disease include tuberculosis, MRSA, VRE, and deep mycosis.
- a foreign gene is further introduced as described later, an effect is expected for various gene diseases.
- the T cell population produced by the method of the present invention can also be used for donor lymphocyte infusion for the purpose of bone marrow transplantation, prevention of infection after irradiation, and remission of relapsed leukemia.
- the present invention expresses CD45RA obtained by the method for producing a T cell population of the present invention, and expresses at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28.
- a T cell population is provided.
- the present invention also provides a medicament (therapeutic agent) containing the T cell population as an active ingredient.
- the therapeutic agent containing the T cell population is suitable for use in immunotherapy.
- immunotherapy T cells suitable for treatment of a patient are administered to a patient by an administration method such as intravenous, arterial, subcutaneous, or intraperitoneal injection.
- the therapeutic agent is very useful for use in the aforementioned diseases and donor lymphocyte infusion.
- the therapeutic agent is in accordance with a known method in the pharmaceutical field, for example, using the T cell population prepared by the method of the present invention as an active ingredient, a known organic or inorganic carrier suitable for parenteral administration, excipient, stable It can be mixed with pills and prepared as infusions or injections.
- a known organic or inorganic carrier suitable for parenteral administration, excipient, stable It can be mixed with pills and prepared as infusions or injections.
- the content of the T cell population of the present invention in the therapeutic agent, the dosage of the therapeutic agent, and various conditions relating to the therapeutic agent can be determined as appropriate according to known immunotherapy.
- the content of the T cell population of the present invention in medicine is not particularly limited, but for example, preferably 1 X 10 3 to 1 X 10'ells / mL, more preferably 1 X 10 4 to Examples are 1 ⁇ 10 10 cells / mL, more preferably 1 ⁇ 10 5 to 1 ⁇ 10 9 cells / mL.
- the dose of the medicament of the present invention is not particularly limited. For example, it is preferably 1 X 10 6 to 1 X 10 12 cells / day, more preferably 1 X 10 7 to Examples are 5 X lOUcells / day, more preferably 1 X 10 8 to 2 X lC ⁇ cellsZ days.
- immunotherapy with the therapeutic agent is used in combination with known drug administration, radiotherapy, and surgical treatment. Say it with a word.
- the method for producing a T cell population of the present invention can further include a step of introducing a foreign gene into the sputum cells. That is, the present invention provides, as one aspect thereof, a method for producing a sputum cell population that further includes a step of introducing a foreign gene into the sputum cell.
- the term “foreign gene” refers to a gene that is artificially introduced into a target cell for gene transfer, and includes those derived from the same species as the target cell for gene transfer.
- the means for introducing a foreign gene there are no particular limitations on the means for introducing a foreign gene, and an appropriate one can be selected and used by a known gene introduction method.
- the process of gene transfer can be performed at any time during the production of the sputum cell population.
- the ability to be performed simultaneously with, during, or after the production of the sore cell population is also suitable from the viewpoint of work efficiency.
- the viral vector is not particularly limited, and is usually a known winores betater used in gene transfer methods, for example, retro winores betater, lentiwinores vector, adenovirus vector, adeno-associated virus vector, Simian virus vectors, silkworm your virus vectors, Sendai virus vectors, etc. are used.
- a retrovirus vector an adenovirus vector, an adeno-associated virus vector, a lentivirus vector or a simian virus vector is used.
- the above-mentioned virus vector those lacking the replication ability are preferable so that they cannot self-replicate in infected cells.
- a substance that improves gene transfer efficiency such as RetroNectin (registered trademark, manufactured by Takarabio Co., Ltd.) can also be used during gene transfer.
- the retrovirus vector and the lentivirus vector are stably inserted into the chromosomal DNA of the cell into which the vector is introduced, and the foreign gene is stably integrated. It is used for gene therapy and other purposes. Since the vector has high infection efficiency with respect to dividing and proliferating cells, it is suitable for gene introduction in the production process of the present invention.
- the method for gene transfer without using a viral vector is not limited to the present invention.
- a method using a carrier such as ribosome or ligand-polylysine, a calcium phosphate method, an electopore position method, The particle gun method can be used.
- a foreign gene incorporated into plasmid DNA, linear DNA or RNA is introduced.
- the foreign gene introduced into the T cell there are no particular limitations on the foreign gene introduced into the T cell, and any gene desired to be introduced into the cell can be selected. As such a gene
- antisense nucleic acids eg, enzymes, cytodynamics, receptors, etc.
- siRNA small interfering RNA
- ribozyme coding can be used.
- an appropriate gene that enables selection of the transfected cells may be introduced simultaneously.
- the foreign gene can be used by inserting it into a vector or a plasmid so that it can be expressed under the control of an appropriate promoter, for example.
- an appropriate promoter for example.
- other regulatory elements cooperating with the promoter and transcription initiation site for example, an enhancer sequence or a terminator sequence may be present in the vector.
- an enhancer sequence or a terminator sequence may be present in the vector.
- each base sequence on both sides of the desired target insertion site of the gene in the chromosome is homologous.
- An exogenous gene may be arranged between flanking sequences consisting of the nucleotide sequences.
- the foreign gene to be introduced may be a natural gene, an artificially produced gene, or a DNA molecule having a different origin and bound by a known means such as ligation. Furthermore, it may have a sequence in which a mutation is introduced into a natural sequence according to the purpose.
- a gene encoding an enzyme related to resistance to a drug used for treatment of a patient such as cancer is introduced into a T cell to impart drug resistance to the T cell.
- a combination of immunotherapy and drug therapy Therefore, a higher therapeutic effect can be obtained.
- drug resistance genes include multidrug resistance genes.
- a gene that confers sensitivity to a specific drug can be introduced into T cells to impart sensitivity to the drug.
- T cells after transplantation into a living body can be removed by administration of the drug.
- An example of a gene that confers sensitivity to a drug is a thymidine kinase gene.
- genes to be introduced include a gene encoding TCR that recognizes the surface antigen of the target cell, an antigen recognition site of an antibody against the surface antigen of the target cell, and an intracellular region of TCR (CD3 Etc.) and the like.
- the present invention also provides a method for treating or preventing a disease, comprising administering to a subject an effective amount of a T cell population obtained by the aforementioned method.
- the subject is not particularly limited, but preferably refers to a patient having a disease as described above, to which a T cell population produced by the method of the present invention is administered.
- an effective amount means that when the T cell population is administered to the subject, treatment or treatment is performed compared to a subject not administered with the T cell population.
- the specific effective amount is appropriately set according to the administration form, administration method, purpose of use and age, weight, symptom, etc. of the subject.
- the administration method is not limited. For example, it may be administered by drip, injection or the like, as in the case of the above-mentioned medicine.
- the present invention also provides the use of the aforementioned T cell population for the manufacture of a medicament.
- the method for producing the medicament is performed in the same manner as the aforementioned medicament.
- the disease to which the drug is administered is not particularly limited, but is the same as the aforementioned drug.
- the present invention provides a T cell that expresses CD45RA and expresses at least one selected from the group consisting of CD62L, CCR7, CD27, and CD28 obtained by the production method of the present invention described above. From the group consisting of cells capable of presenting antigens to the population, cells presented with antigens, cells capable of producing antigens, CD3 ligands, CD28 ligands, cytoforce-ins, chemokines and cytokines At least one stimulator selected By applying the stimulation, an activated ⁇ cell population can be produced. Furthermore, the present invention provides a ⁇ cell population obtained by the above production method.
- the activated T cell population obtained in this way can be used as an active ingredient of a medicine in the same manner as the T cell population obtained by the production method described above.
- the stimulation by the stimulating factor is not particularly limited as long as the T cell population obtained by the above-described production method of the present invention is activated by the stimulating factor, but for example, by the production method of the present invention. It is exemplified that the culture is carried out in the coexistence of the obtained ⁇ cell population and the stimulating factor.
- a cell having an ability to present an antigen is not particularly limited as long as it is a cell generally used as an antigen-presenting cell.
- a rod-shaped cell, a ⁇ ⁇ ⁇ cell, a single cell can be used.
- Examples include spheres, sputum cells, sputum cells, macrophages, fibroblasts, Langerhans cells, and cell populations containing at least one of these cells.
- Particularly preferred are spider cells, ⁇ cells, Examples are sputum cells, sputum cells, monocytes, macrophages, and cell populations containing at least one of these cells.
- the origin of the cell having the ability to present the antigen may be either self or non-self for the patient to be administered, but the self-derived one is preferably used.
- a cell having an ability to present an antigen means a cell having an ability to present an antigen but not presenting an antigen.
- the antigen-presented cell is a cell in which an appropriate antigen is artificially added to a cell having the ability to present the antigen, or already collected when collected from a living body. It can also be used to present and shift cells.
- the cell on which the antigen is presented can be produced and used in the same manner as described in Example 5- (5) below.
- “cells having the ability to present an antigen” are not included in the meaning of the phrase “cells presented with an antigen”.
- the antigen is not particularly limited as long as the peptide is presented on the antigen-presenting cell and can be recognized by the cell and can efficiently activate the cell, for example, the peptide And glycopeptide, tumor cell extract, tumor cell sonication product and tumor cell hydrothermal treatment product, virus, bacteria, protein and the like.
- CD3 ligand and CD28 ligand the CD3 ligand described above is used.
- Doya CD28 ligand is exemplified.
- CD45RA obtained by the above-described production method of the present invention is expressed, and at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28 is expressed.
- cyto force-in is not particularly limited as long as it acts on T cells and can be activated.
- IL-2, IFN-y, TGF- ⁇ , IL 15, IL-7, IFN- ⁇ , IL-12, CD40L, IL-27, etc. are exemplified, and the viewpoint power to enhance cellular immunity is particularly preferably exemplified by IL-2, IFN- ⁇ , IL-12 .
- the chemokine is not particularly limited as long as it acts on T cells and exhibits migration activity.
- RANTES, CCL21, MIP1 a, MIP1 ⁇ , CCL19, CX CL12, IP-10 MIG is exemplified.
- the cell having the ability to produce cytokines is not particularly limited as long as it has the ability to produce the above-mentioned cytokines.
- Thl cells are preferably used from the viewpoint of enhancing the activity.
- the T cell population obtained by the production method of the present invention described above is undifferentiated and subjected to antigen stimulation! /, Because it contains a high ratio of naive T-like cells, the stimulation used here As a factor, it is particularly preferable to use a cell having an ability to present an antigen, a cell on which an antigen is presented, and Z or an antigen. For example, as shown in Examples 5— (5), 6— (8), 7— (2), 8— (1), 14 (7) and 15— (2) described later, the production of the present invention described above is performed.
- antigen stimulation to the T cell population obtained by the method, it is possible to induce useful antigen-specific CTLs with extremely high cytotoxic activity and high antigen recognition ability.
- the culture can be performed in the presence of the aforementioned fibronectin, a fragment thereof, a mixture thereof, or a known component used for T cell culture.
- the T cell population produced in this way is a highly useful T cell population with a high therapeutic effect.
- the present invention also provides a method for treating or preventing a disease, comprising administering to a subject an effective amount of an activated T cell population obtained by the aforementioned method.
- this departure Ming also provides the use of the aforementioned activated T cell population for the manufacture of a medicament.
- the method for producing the medicament is performed in the same manner as the aforementioned medicament.
- the disease to which the drug is administered is not particularly limited, but is the same as the aforementioned drug.
- the present invention provides (a) a CD45RA obtained by the production method of the present invention described above and expressing at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28.
- a pharmaceutical comprising a formulation containing at least one selected stimulator as an active ingredient, wherein the formulation is contained as two separate formulations administered simultaneously or separately.
- those that can give antigen stimulation that is, those that can be used as vaccines, for example, cells capable of presenting antigens, cells that present antigens, antigens, CD3 ligands, CD28 ligands, sites Cell force having the ability to produce force-in, chemokine and cyto force-in At least one selected from the group consisting of cells capable of presenting a preferred antigen, a cell presented with an antigen, or an antigen It is preferably used according to the invention.
- the T cell population that expresses (45) CD45RA obtained by the production method of the present invention and expresses at least one selected from the group consisting of CD62L, CCR7, CD27, and CD28 in the medicament is an active ingredient
- the content, dosage, and mode of administration of a T cell population that expresses CD45RA in the drug and expresses at least one selected from the group consisting of CD62L, CCR7, CD27, and CD28 are, in particular, although there is no limitation, for example, it can be the same as the pharmaceutical containing the T cell population obtained by the production method of the present invention described above.
- a cell capable of presenting an antigen, an antigen-presented cell, an antigen, a CD3 ligand, a CD28 ligand, a cyto force-in, a chemokine, and a cytokin in the medicament are produced.
- a preparation containing, as an active ingredient, at least one stimulating factor selected from the group consisting of capable cell forces for example, a preparation prepared by combining the stimulating factor with an appropriate carrier can be used.
- a cell having an ability to present an antigen a cell having an antigen presented, an antigen, a CD3 ligand, a CD28 ligand, a cyto force in, a chemokine and a cell force having an ability to produce a site force in are selected from the group consisting of As a specific example of at least one stimulation factor, for example, those exemplified above can be used.
- a cell having the ability to present an antigen as a stimulating factor a cell on which an antigen is presented
- the cells When cells having the ability to produce an antigen or cytodynamic in are used, the cells can be formulated in combination with an appropriate infusion or injection pharmaceutical carrier, although there is no particular limitation. Moreover, when using an antigen peptide, although there is no limitation, it can combine with a suitable adjuvant. When cytokine chemokine is used, it can be formulated so as to be incorporated into ribosomes by a known method.
- the content of the stimulating factor in the preparation can be appropriately selected depending on the type of the stimulating factor to be used, and is not particularly limited. For example, cells having the ability to present an antigen as the stimulating factor, antigen presentation When using cells that have been made or have the ability to produce cyto force-in
- the administration form of the stimulating factor can be appropriately selected depending on the type of the stimulating factor used, and is not particularly limited.
- intravenous administration, intraarterial administration, subcutaneous administration, intraperitoneal administration, oral administration, etc. Be administered.
- the dose is not particularly limited as long as it is an amount effective for treating or ameliorating a patient's disease.
- a cell having the ability to present an antigen is administered, for example, it is preferably used.
- 1 X 10 3 to 1 X 10 cells / day more preferably 1 X 10 3 to 1 X 10 10 cells / day, more preferably 1 X 10 4 to 1 X 10 9 cells / day Is administered, for example, preferably from 0.001 to 100 mg / day, more preferably from 0.003 to 30 mgZ day, particularly preferably from 0.01 to: LOmgZ day.
- administration of the preparation (b) for example, it is preferable to administer, as a preparation (b), a combination of cells capable of presenting an antigen and an antigen.
- a combination of cells capable of presenting an antigen and an antigen For example, it is preferable to administer the cells in combination with antigenic peptides.
- the contents and doses of the cells having the ability to present the antigen and the antigen in the preparation can be carried out as described above.
- the administration of the preparation (a) and the preparation (b) in the medicine is a force that is administered to the patient at the same time or separately.
- the preparation (a) and the preparation (b) are administered as an infusion
- an embodiment in which these preparations are mixed and administered to a patient before administration is included in the present invention.
- “separately” means that the preparation (a) and the preparation (b) are administered separately in terms of time.
- the administration interval between the preparation (a) and the preparation (b) There is no particular limitation as long as the stimulation of the stimulating factor contained in the preparation (b) can be applied to the T cell population contained in the preparation (a) in the body, but preferably (a) It is preferable to administer the preparation (b) after administering the preparation. There are no particular restrictions on the number of doses, and the dose can be administered once or divided into several doses.
- the present invention provides (a) a CD45RA obtained by the production method of the present invention described above and expressing at least one selected from the group consisting of CD62L, CCR7, CD27 and CD28.
- Administering a cell population to a patient and (b) producing cells capable of presenting an antigen, cells presented with an antigen, antigen, CD3 ligand, CD28 ligand, cytoforce-in, chemokine, and cytoforce-in
- a method of treating a disease comprising the step of administering to a patient at least one stimulating factor selected from the group consisting of the cellular forces having the ability to act.
- the treatment method can exhibit an extremely high therapeutic effect.
- a stimulating factor those that can provide antigen stimulation, that is, those that can be used as vaccines, for example, cells that have the ability to present antigen, cells that present antigen, antigen, CD3 ligand, CD28 At least one selected from the group consisting of a ligand, a cytodynamic force, a chemokine, and a cellular force capable of producing a cytodynamic force can present a preferred antigen.
- a cell having capacity, a cell on which an antigen is presented, or an antigen is preferably used according to the present invention.
- the step of administering the T cell population obtained by the method of the present invention to a patient is not particularly limited, but for example, the same administration as the above-mentioned pharmaceutical administration method
- the dosage and administration form can be adopted.
- cells having the ability to present antigen, cells presented with antigen, antigen, CD3 ligand, CD28 ligand, cytokine, chemokine and cell force capable of producing cyto force-in The step of administering the selected at least one stimulating factor to the patient is not particularly limited, and can be carried out, for example, by adopting the same dosage and administration form as those of the aforementioned medicament.
- the administration interval between the step (a) and the step (b) can be performed in the same manner as the preparation containing the preparation (a) and the preparation (b) described above. There is no particular limitation as long as the stimulation of the stimulating factor administered in the step (b) can be applied to the T cell population administered in the step (a), but it is performed simultaneously or separately. Preferably, the administration step (b) is preferably performed after the administration step (a).
- the number of doses is not particularly limited, and can be administered once or divided into several times.
- the blood sample was diluted 2-fold with phosphate buffered saline (hereinafter referred to as PBS) and Ficoll-paque (Amersham Bioscience)
- PBS phosphate buffered saline
- Ficoll-paque Amersham Bioscience
- the product was overlaid on the product and centrifuged at 600 ⁇ g for 20 minutes.
- Terminal blood mononuclear cells hereinafter referred to as PBMC
- the collected PBM C is a stock solution consisting of 90% FBS (Cambrex) ZlO% DMSO (Sigma)!
- HSA human serum albumin
- CP Suspended in a stock solution consisting of an equal mixture of 1 (manufactured by Kyokuto Yakuhin Co., Ltd.) and RPMI1640 medium (manufactured by Sigma) and stored in liquid nitrogen. Store these PBMCs in a 37 ° C water bath for T cell expansion. After rapid thawing and washing with RPMI1640 medium containing 10 / z gZmL DNase (Calbiochem), the number of viable cells was calculated by trypan blue staining method and used for each experiment.
- HSA human serum albumin
- Anti-human CD3 antibody and CH-296 fragment were immobilized on the culture equipment used in the following experiments. Specifically, 1.9 mL of PBS containing anti-human CD3 antibody (manufactured by Janssen Pharma) (final concentration 5 / z gZmL) was added to a 12-well cell culture plate (Betaton Dickinson). At this time, CH-296 was added to the CH-296 addition group so as to have a final concentration (25 / z gZmL).
- Example 1- (1) Prepared in Example 1- (1) so that AIM-V containing 1% HumanAB serum (Cambrex) (Invitrogen, hereinafter abbreviated as 1% AIM-V) was 1 ⁇ 10 6 cellsZmL.
- 1% AIM-V HumanAB serum
- the cell solution was added with 2 mL LZ well, and 1 mL Zwell was added to each cell solution.
- IL-2 Proleukin: manufactured by Chiron
- Example 1 Cells prepared at each culture date and time in Example 1 (3) were washed with PBS and PBS containing 1% bovine serum albumin (Sigma, hereinafter referred to as 1% BSAZPBS). The cells were suspended in 1% BSAZPBS, and FITC-labeled mouse IgG 1ZRD1-labeled mouse IgG 1ZPC5-labeled mouse IgG 1 (manufactured by Beckman Coulter) was added as a negative control. Similarly, FITC-labeled mouse anti-human CD62L antibody (manufactured by Sigma) ZRD1-labeled mouse anti-human CD45RA antibody (manufactured by Beckman Coulter) was added.
- BSA bovine serum albumin
- Example 1 After preparing the cell solution by suspending the PBMC prepared in Example 1 (1) so that it becomes 1 X 10 6 cells ZmL in AIM-V containing 3% Human AB serum (hereinafter abbreviated as 3% AIM-V)
- Example 1 add 3% AIM-V to the anti-human CD3 antibody-immobilized plate prepared in (2) or anti-human CD3 antibody and CH-296-immobilized plate with 2 mL Zwell, and The cell fluid was added in lmLZ wells. IL-2 was added to a final concentration of lOOOUZmL, and these plates were cultured at 37 ° C in 5% CO (culture day 0). On the 4th day
- IL-2 was added to each group to a final concentration of 500 UZmL.
- the number of viable cells was counted by the trypan blue staining method, and the expansion culture rate was calculated by comparison with the number of cells at the start of culture. The results are shown in Table 3.
- Serum concentration (%) Number of days of culture Magnification rate (magnification) Control (C H-2 9 6 non-immobilized) 3 ⁇ 1 ⁇ 0.0 5 1 4 days X 2 7 6
- Example 2 Cells prepared in (1) were washed with PBS and 1% BSAZPBS. The cells were suspended in PBS containing 1% BSA, and FITC-labeled mouse IgG1 / R D1-labeled mouse IgGlZPC5-labeled mouse IgGl + ECD-labeled mouse IgGl (manufactured by Beckman Coulter, Inc.) was added as a negative control. Similarly, RD1-labeled mouse anti-human CD45RA antibody ZFITC-labeled mouse anti-human CCR7 antibody (manufactured by R & D Systems) ZECD-labeled mouse anti-human CD8 antibody (manufactured by Beckman Coulter) was added.
- CD 8 Spider cells (CD 4+ T cells) CD 45 RA— CCR 7 + T cells (%) Control (CH—296 unfixed) 9.7
- Example 3 Analysis of cytoforce-in productivity of CD45RA + CCR7 + T cells and CD45RA-CCR7-T cells in a T cell population expanded using CH-296
- Example 1 After suspending PBMC prepared in 1 (1) and preparing a cell solution, anti-human CD3 antibody-immobilized plate prepared in Example 1 1 (2), or anti-human CD3 antibody and CH-296 immobilized 0.5% GT-T503 was added to the cell with 0.5 mL Zwell, and the above cell solution was added to each lmLZ well. IL-2 was added to a final concentration of lOOOUZmL, and these plates were cultured at 37 ° C in 5% CO (culture day 0). On day 4 of culture start
- CH-296 group As shown in Table 6, in the group using CH-296 fixed in the initial stage of T cell population expansion culture (hereinafter referred to as CH-296 group), T The expansion rate of the cell population was high.
- Example 3- (1) The cells prepared in Example 3- (1) were stained with each antibody and analyzed in the same manner as in Example 1-1 (4). However, antibody combinations were performed as follows. Specifically, FIT C-labeled mouse IgG1 / RD1-labeled mouse IgG1 ZPC5-labeled mouse IgG1 and RD1-labeled mouse anti-human CD45RA antibody ZFITC-labeled mouse anti-human CCR7 antibody was stained. These were analyzed with a flow cytometer, and the ratio of CD45RA + CCR7 + T cells was calculated. The results are shown in Table 7.
- Example 3- After the cells prepared in (1) were stained by the same method as in Example 2- (2), the cells were washed with 1% BSAZPBS and suspended in GT-T503 medium. These cells were transferred to CD8RA + CCR7 + T cells and CD45RA ⁇ CCR7 ⁇ T cells using CD8RA + CCR, which is the majority of CD4 + T cells. The cells were sorted into 7+ T cells and CD45RA-CCR7-T cells. The same stained cells before sorting are subjected to flow cytometry, and the CD8 + T cell region and CD The cells were classified into 8_T cell regions, and the percentage of CD45RA + CCR7 + T cells was calculated for each cell population. The results are shown in Table 8 and Table 9.
- CD 45 RA 'CCR 7+ T cells (3 ⁇ 4;)
- Anti-human CD3 antibody and anti-human CD28 antibody were immobilized on the culture equipment used in the following experiments. That is, add 96 L of acetate buffer ( ⁇ 5.3) containing anti-human CD3 antibody (2 gZmL) to a 96-well cell culture plate (Betaton Dickinson), and add anti-human CD 28 antibody (Dako). 80 ⁇ L of acetate buffer containing 20 ⁇ g / mL was added. The final concentration of anti-human CD3 antibody was 1 ⁇ g / mL, and the final concentration of anti-human CD28 antibody was 10 ⁇ g ZmL. These culture devices were incubated at room temperature for 5 hours and then stored at 4 ° C until use. Immediately before use, the buffer solution containing these antibody was removed by aspiration, and each well was washed twice with PBS and once with GT-T503 medium for the experiment.
- Naive T cells and central memory T cells are known to produce a large amount of IL-2 when antigen-stimulated in the body.
- effector memory T cells produce a large amount of IFN- ⁇ and IL-4 upon antigen stimulation.
- Example 3 In (3) In order to confirm whether or not each of the obtained cell fractions maintained these functions, the ability to produce cyto force when stimulated with anti-CD3 antibody and anti-CD28 antibody was measured.
- Example 3 Each cell population obtained in (3) was collected and then suspended in 0.5% GT-T503 to count the number of cells.
- Example 3 Cells were added to each well so as to be 2 ⁇ 10 5 cells / 0.2 mL on the anti-human CD3 antibody and anti-human CD28 antibody-fixed antibody plate prepared in (4), and 37 ° C. in 5% CO. At C
- Example 3- (3) in the CH-296 group, the CD 45RA + CCR7 + T cell ratio was higher V than in the control group, and the results were obtained.
- the CD45RA + CCR7 + T cell population retains its properties as a naive T-like cell
- the CD45RA + CCR7 + T cells and CD45RA-CCR7- T cells isolated after expansion were expanded. Site power in productivity was evaluated.
- IL-2 and IFN-y are produced using ELISA Development Kit (R & D Systems), and IL-4 is produced by READY-SET-GO! Measurement was performed using Human Interleukin-4 (manufactured by eBioscience).
- Table 10 and Table 11 show the results of each site force-in production in the control group and the CH-296 group, respectively. 1
- T cells CD45RA CCR 7 Spider cells 104. 9 1229. 0 430. 8
- CD 8 "CD 45 R ⁇ + CCR 7 + ⁇ cells 1 5; ⁇ 9.4 1 07. 9 1 13. 9
- T cells CD 45 KA "CCR 7 ⁇ cells 61 1. 0 587. 5 727. 3 [0139] As shown in Tables 10 and 11, IL-2 production was also confirmed in the control group and the CH-296 group. In both groups, CD8-T cells produced more than CD8 + T cells, and CD45 RA + CCR7 + T cells produced more than CD45RA-CCR7-T cells. From this, it was shown that the CD45RA + CCR7 + T cell population obtained by T cell population expansion culture has properties as na ⁇ ve T-like cells.
- IFN- ⁇ and IL-4 are produced predominantly in CD45RA-CCR7-T cells in the control group and CH-296 group, and the CD45RA-CCR7-T cell population maintains the effector memory function. It was suggested to be a group.
- Example 1 Performed in the same manner as in (2). However, PBS containing anti-human CD3 antibody (final concentration 5 ⁇ g / mL) was added in an amount of 0.45 mL each.
- CH-296 group T cells were compared with the control group.
- the expansion rate of the population was high.
- Example 4 The cells prepared in (1) (2) were treated in the same manner as in Example 3-(2) using the CD45RA + CC The ratio of R7 + T cells was calculated. The results are shown in Table 13.
- CCR7 positive cells such as naive T cells and central memory T cells react to the chemokine CCL21 and show chemotaxis. This is an important event for these cells to migrate from blood vessels to lymph nodes, and it was confirmed that the cells obtained after expansion culture had chemotaxis to CCL21.
- reaction medium RPMI164 0 medium (hereinafter referred to as reaction medium) containing 0.5% BSA to give 5 ⁇ 10 6 cellsZmL. .
- Example 4 Performed in the same manner as in (1).
- Example 4 Performed in the same manner as in (2). The results of the expansion culture rate are shown in Table 15.
- Example 5 Cells prepared in (2) were stained with each antibody and analyzed in the same manner as in Example 1— (4). However, antibody combinations were performed as follows. Specifically, FI TC-labeled mouse IgGlZRDl-labeled mouse IgGl (all manufactured by Dako), RD1-labeled mouse anti-human CD45RA antibody ZFITC-labeled mouse anti-human CCR7 antibody and RD1-labeled mouse anti-human CD45RA antibody ZFITC-labeled mouse anti-human CD62L antibody (this Thereafter, the anti-human CD62 L antibody was stained with eBioscience. These were analyzed by a flow cytometer, and the ratio of CD45RA + CCR7 +, CD45RA + CD62L + T cells was calculated. The results are shown in Table 16. [0156] [Table 16] Table 1 6
- the CH-296 group showed a high value for any cell surface marker as compared to the control group.
- Example 5 Cultured 14th day cells prepared in (2) were suspended in a stock solution consisting of an equal mixture of CP-1 and RPMI1640 medium containing 90% FBSZlO% DMSO or 8% HSA, and liquid nitrogen Saved in. When CTL is induced, these preserved cultured cells are rapidly thawed in a 37 ° C water bath, washed with RPMI1640 medium containing 10 / z gZmL DNase, and the number of viable cells is calculated by trypan blue staining method and used for each experiment. did.
- Example 5 Using the cells prepared in (4), anti-tumor-associated antigen (Melanoma antigen ecologized by Tcells, MART-1) -specific CTL was induced. Induction of anti-tumor-associated antigen (MART-1) -specific CTL is performed by the method of Prebanski M. et al. [Plebanski M. et al., Eur. J. Immunol., 25th, No. 6, 1783-1787. Page (1995)] was partially modified.
- PBMC prepared in Example 1 1 (1) was used as an antigen-presenting cell, and 40 g / mL of melanoma antigen MART-1-derived epitope peptide (the melanoma antigen MART described in SEQ ID NO: 22 in the Sequence Listing) was used as the antigen peptide.
- 1 HLA-A2. 1 binding 3 ⁇ 4 peptide) and 3 ⁇ g / mL ⁇ -microglobulin (Scrips)
- RPMI1640 medium (hereinafter abbreviated as 5HRPMI) containing 5% Human AB serum, 0. ImM NEAA mixture ⁇ ImM Sodium pyru vate, 2 mM L-glutamine (all manufactured by Cambrex), 100 gZmL streptomycin (Meiji Seika Co., Ltd.) ) In a 5% CO incubator at 37 ° C for 2 hours. After incubation, X
- Example 5- (4) the cultured cells prepared in Example 5- (4) were suspended in 5HRP Ml to 2 ⁇ 10 6 cellsZmL, and 0.5 mLZ was added to a 24-well cell culture plate (Betaton Dickinson). Ye Added in portions. Add 0.5 mLZ of antigen-presenting cells prepared by the above method to each well, and add IL-7 (R & D Systems) and KLH (Calbiochem) to a final concentration of 25 ngZmL, 5 The mixture was added to give gZmL. 5% CO in the plate
- Responder cells cultured for 1 week are suspended in 5HRPMI so as to obtain 1.8 to 2.0 X 10 6 cellsZmL.
- 5HRPMI 0.5 mLZ well was added to each tube, and IL-7 was added at a final concentration of 25 ngZmL, followed by re-stimulation.
- 1 mL of 5HRPMI containing 60 U / mL IL-2 was added to each well.
- day 11 of culture after suspending the cells in each well, divide each half into 2 wells, add 5 mL of 5HRPMI containing 60 U / mL IL-2 to each well, and culture until 15 days. Continued feeding.
- Example 5 On the 7th and 14th day after the start of culture, the number of viable cells was measured by the trypan blue staining method on the cells obtained in (5). Was calculated. The results are shown in Table 17.
- Example 5 The cytotoxic activity of CTL 15 days after the start of induction prepared in (5) was determined by measuring the cytotoxic activity using Calcein-AM [Lichtenfels R. et al. ), J. Immunol. Methods, No. 172, No. 2, pp. 227-239 (1994)].
- the HLA-A2.1-retaining cell line T2 cells ATCC CRL-1992
- Calcein-AM Dojindo Laboratories
- the cells were washed with a medium not containing Calcein-AM and then used as Calcein-labeled target cells.
- Calcein-labeled target cells were mixed with 30 times the amount of K562 cells (Human Science Research Resource Bank JCRB0019) to prepare cells for measuring cytotoxic activity.
- K562 cells were used to eliminate non-specific damage activity by NK cells mixed in responder cells.
- Example 5 Using CTL prepared in (5) as effector cells, serially dilute with 5HRPMI to 3 ⁇ 10 5 to 3 ⁇ 10 6 cells / mL, and add 100 to each well of a 96-well cell culture plate. Aliquots of ⁇ LZ wells were added, and 100 LZ wells were added to the cells for measuring cytotoxic activity prepared so that the Calcein-labeled target cells were 1 X 1C ZmL. At this time, the ratio of the effector cells (E) to the Calcein-labeled target cells (T) was shown as the EZT ratio, and the EZT ratios 30, 10, and 3 were measured. Centrifuge the plate containing the cell suspension at 400 8 minutes for 1 minute, and incubate at 37 ° C for 4 hours in a 5% CO incubator.
- the minimum release amount is only for cytotoxic activity measurement cells This is the amount of calcein released from the contained well, and the amount of spontaneous release of calcein from calcein-labeled target cells.
- the maximum released amount indicates the amount of Calcein released when the cells are completely destroyed by adding 0.1% surfactant Triton X-100 (manufactured by Nacalai Tester) to the cells for measuring cytotoxic activity! .
- the results of the cytotoxic activity measurement are shown in Table 18.
- Example 6 Induction of Allogeneic MLR and anti-MART-1 CTL in expanded cell populations using a gas-permeable culture bag after initial stimulation of CH-296
- Anti-human CD3 antibody and CH-296 were immobilized on the culture equipment used in the following experiments. Specifically, 9 mL of PBS containing anti-human CD3 antibody (final concentration 5 g / mL) was added to each 75 cm 2 cell culture flask (Betaton Dickinson). At this time, CH-296 was added to the CH-296 addition group so that the final concentration (25 gZmL) was obtained.
- CH- 296 group 6
- CH- 296 group 6
- Example 6 Cells prepared in (2) were stained with each antibody and analyzed in the same manner as in Example 1— (4). However, antibody combinations were performed as follows.
- CD45 RA + CD 28 + T cells 36.9% 6 3.0%
- the CH-296 group showed higher values for all cell surface markers than the control group.
- CD27 and CD28 are known to have a high expression rate in undifferentiated cells such as na ⁇ ve T cells. Even when analyzed with these markers, T- It became clear that the na ⁇ ve T-like cell population ratio in the cell population was increased.
- Example 6 Allogeneic MLR was performed using the cells prepared in (2). That is, Example 1— PBMC derived from an Allogeneic donor (non-self donor: a donor different from the donor used in Example 6-(2)) prepared by the same method as (1) was irradiated with X-rays (0.88C / kg), and 5 HRPMI was prepared to 2 ⁇ 10 6 cells / mL (Stimulator cell). On the other hand, the cultured cells prepared in Example 6- (2) were suspended in 5HRPMI at 2 ⁇ 10 6 cells / mL (Responder cell). Stimulator cells and Responder cells prepared on 24-well cell culture plates were added to each 0.5 mL Z well. A final concentration of 500 for each well After adding IL-2 to UZmL, plate at 37 ° C in a 5% CO incubator.
- Example 6 On the 7th and 10th day after the start of culture, the number of viable cells was measured by the trypan blue staining method, and the expanded culture rate was compared with the number of cells at the start of the culture. Was calculated. The results are shown in Table 21.
- Example 6 The cytotoxic activity of the cells on day 10 after the start of induction prepared in (4) was performed in the same manner as in Example 5- (7). However, as target cells at this time, autologous PBMCs or non-self PBMCs blasted with Phytohemagglutin in (hereinafter abbreviated as PHA) for 10 days were used as force lusein-labeled target cells. When measuring the cytotoxic activity, 30 times the amount of K562 cells and force-labeled target cells were mixed. The results of cytotoxic activity measurement are shown in Table 22.
- PHA Phytohemagglutin in
- Example 6 Cultured 14th day cells prepared in (2) were frozen in the same manner as in Example 5— (4).
- Example 6 The cultured cells prepared in (7) were suspended in 5HRPMI to 1 ⁇ 10 6 cells / mL, and 5HRPMI 1mL containing 60UZmL of IL-2 was added to each well on the second day of culture.
- the responder cells are suspended in 5HRPMI so that the responder cells become 0.3 to 1.3X10 6 cellsZmL, and the antigen-presenting cells become 1.6X10 6 ce UsZmL, and the culture starts.
- 5HRPMI lmL containing 60UZmL of IL-2 was added to each well.
- cells in each well were suspended and divided into two wells in half.
- 5HRP MI 1mL containing 60UZmL IL 2 was added to each well. [0188] (9) Measurement of cytotoxic activity
- Example 7 Anti-MART-1CTL induction from expanded cultured cell-derived CD45RA + CCR7 + CD8 + T cells and CD45RA—CCR7—CD8 + T cells
- Example 1 After staining the cells prepared in (1) and Example 6— (7) by the same method as in Example 2— (2), the cells were washed with 1% BSAZPBS, and IMDM medium (Invitrogene) In the product). The cells were subjected to a high-speed cell sorter Moflo, and the CD45RA + CCR7 + CD8 + fraction and the CD45RA-CCR7-CD8 + fraction were each isolated to obtain a fraction having a purity of 92 to 99%.
- IMDM medium Invitrogene
- Example 7 Using the cells prepared in (1), antitumor-related effects were obtained in the same manner as in Example 6 (8). Antigen (MART-1) -specific CTL was induced and the culture was continued for 13 days. However, changes were made in the following points. 5HRPMI containing 60U / mL IL-2 on the 2nd day after the start of culture, in which 0.25mL each of the culture start cells and antigen-presenting cells were added to 48-well culture plates (Betaton's Dickinson). 5 mL was added to each well, and 4 days after the start of culture, half of the culture supernatant was removed, and 0.5 mL containing 60 UZmL of IL-2 was added to each well.
- 5HRPMI containing 60U / mL IL-2 on the 2nd day after the start of culture, in which 0.25mL each of the culture start cells and antigen-presenting cells were added to 48-well culture plates (Betaton's Dickinson). 5 mL was added to
- Example 7 On the cells obtained in (2), the number of viable cells was measured by trypan blue staining on the 13th day after the start of culture, and the expansion culture rate was calculated by comparison with the number of cells at the start of culture. The results are shown in Table 24.
- CD45RA + CCR7 + CD 8 isolated from cultured cells (hereinafter referred to as CH-296 group) using culture equipment with CH-296 fixed in the early stage of T cell population expansion culture.
- CH-296 group CD45RA + CCR7 + CD 8 isolated from cultured cells (hereinafter referred to as CH-296 group) using culture equipment with CH-296 fixed in the early stage of T cell population expansion culture.
- the expansion rate of the CTL population was higher than that in the control group.
- a larger CTL population can be obtained by inducing CTL from CD45RA + CCR7 + T cells contained in a high proportion of cultured cells using CH-296 fixed in the early stage of T cell population expansion culture. It became clear that it would be obtained.
- Example 7 The cytotoxic activity of CTL 13 days after the start of induction prepared in (2) was performed in the same manner as in Example 5- (7).
- Table 25 shows the results of the cytotoxic activity measurement.
- Example 6- (8) Using the cells prepared in Example 1- (1) and 6- (7), induction of anti-tumor associated antigen (MART-1) -specific CTL was performed in the same manner as in Example 6- (8). Continued for 14 days
- Example 8 The antigen recognition ability of CTL prepared on day 14 after initiation of induction prepared in (1) was determined by the method of Valmori D et al .; Valmori D. et al., J. Immunol., Vol. 160, vol. : 1758 (1998)] was partially modified. That is, T2 cells were suspended in RPMI 1640 medium containing 5% FBS so as to be 1 ⁇ 10 6 cells ZmL, and then Calcein-AM was added to a final concentration of 25 M, followed by incubation at 37 ° C. for 1 hour. The cells after incubation were washed with a medium not containing Calcein-AM, and then adjusted to 2 ⁇ 10 5 cells / mL.
- Calcein labeled target cells 9 50 LZ wells were dispensed into each well of a 6-well cell culture plate. 5HRPMI containing 0-20 IX M antigenic peptide (melanoma antigen MART-1-derived epitope peptide) was added to each well containing 50 to 50 L / z LZ. 5% CO
- Example 8 Adjusted with 5HRPMI so that the CTL prepared in (1) was 6 ⁇ 10 6 cells / mL as effector cells, and then added 50 / x LZ wells to each well on the plate. Centrifuge the plate containing the cell suspension at 400 X g for 1 minute, then in a 5% CO incubator
- Acetate buffer ( ⁇ 5.3) was used to immobilize the anti-human CD3 antibody and CH-296 fragment, and diluted about 14 times on the 4th day after the start of culture. to that upright 2 culture flask, except that the culture after 8 days was diluted to approximately twice, and transferred to that dilution 6. 3 ml made a 25 cm 2 culture flask, example 3 I went according to (1). The results are shown in Table 27.
- CH-296 group the group in which the culture equipment in which the CH-296 fragment was immobilized was fixed in the initial stage of T cell population expansion culture was compared with the control group. A high expansion culture rate was obtained.
- Example 9 Cells prepared in (1) were stained with each antibody and analyzed in the same manner as in Example 1— (4). However, antibody combinations were performed as follows. That is, staining was performed with FITC-labeled mouse IgG1 ZRD1-labeled mouse IgG1 ZPC5-labeled mouse IgG1 and FITC-labeled mouse anti-human CCR7 antibody ZRD1-labeled mouse anti-human CD45RA antibody ZPC5-labeled mouse anti-human CD62L antibody. These were analyzed by a flow cytometer, and the ratio of CD45RA + CCR7 +, CD45RA + CD62L + T cells was calculated. The results are shown in Table 28 and Table 29.
- Example 9 The cells after expansion culture obtained in (1) (1) were prepared to 2 ⁇ 10 6 cells Z mL using 5HRPMI, and 0.5 mL Zwell was added to each 24-well cell culture plate.
- autologous PBMC prepared in Example 1- (1) was suspended in 5HRPMI, irradiated with X-rays (0.9 OCZkg), and again prepared to 2X10 6 cellsZmL using 5HRPMI (referred to as feeder cells).
- feeder cells were added to the above-mentioned 24-well cell culture plate in 0.5 mLZ wells and cultured at 37 ° C for 7 days in a 5% CO incubator. During this time, IL-2 etc.
- Example 9 1 (3) The cells obtained in Example 9 1 (3) were collected and stained according to the protocol of Annexin V / 7AAD kit (manufactured by Beckman Coulter, Inc.) in order to measure apoptotic cells. The cells were subjected to flow cytometry, and the ratio of Annex inV + 7AAD + cells, ie, cells that had undergone apoptosis, was calculated for each cell population. The results are shown in Table 30.
- CH-2 96 44.9 Based on the results in Table 30, AnnexinV + 7AAD + cells were compared with the control group in the group in which CH-296 was fixed in the initial stage of expansion of the T cell population compared to the control group. The population was low and the percentage of apoptotic cells was shown to be low. In other words, by using the CH-296 fragment for expansion of T cell populations, for example, even in an environment with low IL-2, It was revealed that high cells can proliferate preferentially.
- Mouse CH-296 represented by SEQ ID NO: 23 in the sequence listing was designed from a mouse fibronectin fragment based on the sequence of human CH-296, and a plasmid was constructed according to a conventional method. Obtained engineeringly. That is, E. coli HB101 carrying the plasmid was cultured and induced for expression. Thereafter, the cells were crushed to prepare a crude protein, and then the target protein was purified with a cation exchange column, an anion exchange column, and a gel filtration column, aseptically filtered, and stored at 80 ° C until use.
- IMC carcinoma (hereinafter referred to as IMC) was transplanted into the abdominal cavity of a female CDF mouse (manufactured by SLC Japan) to make ascites and transplanted to another mouse every 7 days. Ascites fluid on the 7th day after subculture was collected, washed with PBS, and suspended in PBS to 5 ⁇ 10 7 cells / mL. 0.1 mL of this cell suspension was transplanted subcutaneously into the right flank of CDF mice to form solid tumors. After 21 days, the spleen was removed and ground in a RPMI 1640 medium using a glass slide.
- RPMI1640 media Seven RPMI1640 media were collected in a tube, collected as a tube, 45 mL, left on ice for 5 minutes, and transferred to a new tube through a 40-m cell strainer (Betaton Dickinsson). After centrifugation, remove the supernatant and suspend in 2 mL of ACK buffer (0.15 M NH 4 Cl, 0.01 M KHCO, 0. OlmM Na EDTA, pH 7.4) for hemolysis.
- ACK buffer 0.15 M NH 4 Cl, 0.01 M KHCO, 0. OlmM Na EDTA, pH 7.4
- RPMI 1640 medium was added so that the cell suspension became 50 mL. After centrifugation, the supernatant was removed, suspended in 10 mL of RPMI1640 medium, and transferred to a new tube through a cell strainer. Add RPMI1640 medium to a cell suspension strength of Om L, centrifuge and remove the supernatant, then suspend and use in an equal volume of RPMI1640 medium and CP-1 containing 8% HSA. Until stored in liquid nitrogen.
- Incubation equipment used in the following experiments includes anti-mouse CD3 antibody and mouse CH-296 Fixed.
- mouse CH-296 prepared in Example 10- (1) was added to a final concentration of 25 ⁇ g ZmL, and further incubated at room temperature for 5 hours.
- the acetate buffer containing antibody / mouse CH-296 was aspirated and removed from the culture equipment, and each well was washed twice with PBS and once with RPMI 1640 medium for the experiment.
- Example 10 The spleen lymphocytes prepared in (2) were purified using nylon fiber to increase the purity of lymphocytes.
- a 10 mL syringe manufactured by Terumo
- nylon fiber manufactured by Wako Pure Chemical Industries
- This column was equilibrated with RPMI1640 medium containing 10% FBS (Dainippon Pharmaceutical Co., Ltd.) and incubated at 37 ° C for 1 hour in a 5% CO incubator.
- Example 10 Prepared in (2)
- mRPMI medium RPMI1640 medium
- Example 10 After suspending the lymphocytes prepared in (4), add mRPMI medium to the anti-mouse CD3 antibody and mouse CH-296 immobilized plate prepared in Example 10- (3) at 0.7 mL Zwell. Each cell solution was added with 0.5 mL Zwell and cultured at 37 ° C in a 5% CO incubator (culture 0
- Example 10 A 6-week-old female CDF mouse was transplanted under the anesthesia under the right flank skin in the same manner as in Example 10- (2).
- Example 10 The cells prepared in (5) were suspended in PBS so as to be 3 ⁇ 10 8 cells ZmL, and 0.1 mL was administered from the tail vein (day 0 of administration). On day 0 of administration, mouse IL-2 (3 ⁇ 10 4 UZ0.2 mL) was administered into the abdominal cavity for 4 consecutive days. As a control, a group not administered with expanded cells was set. The size of the tumor was expressed as a tumor area (cm 2 ) by measuring the major axis and minor axis periodically until 21 days after IMC transplantation. The result is shown in figure 2.
- Figure 2 shows the number of days and tumor size after transplanting IMC.
- the black triangle indicates the control group and the black circle indicates the T cell administration group. As shown in FIG. 2, it was confirmed that tumor formation was significantly suppressed in the group administered with cells cultured in the presence of CH-296 (T cell administration group
- the culture equipment was changed. Specifically, 21 mL of PBS containing anti-human CD3 antibody (final concentration 5 ⁇ g / mL) was added to a 175 cm 2 culture flask (Betaton Dickinson). At this time, CH-296 was added to the CH-296 supplemented caro group to a final concentration of 25 gZmL and incubated at room temperature for 5 hours. Immediately before use, PBS containing the antibody 'CH-296 was aspirated and removed, and each flask was washed twice with PBS and once with RPMI medium. Fresh PBMCs were also prepared for 54 mL blood collection from healthy human donors.
- NODZscid mice manufactured by CLEA Japan, Inc. 8 weeks old, 10 females were weighed and divided into 4 groups.
- the group configuration in this example was as follows.
- Group B PBL + Feeder 1 "h Human IL 2 + Anti-Asialo GM 1 Antibody
- Group C Cells after expanded culture (CH-296 non-immobilized) + Feeder + Hi HL-2 + Anti-ash mouth GM1
- Group D Cells after expansion (CH-296 immobilized) + Feeder + Human IL-2 + Anti-asharo GM1
- Anti-asharo GM1 antibody treatment is known to remove NK cells and enhance human cell engraftment in NODZscid mice. The day before administration of the expanded cells, anti-asharo G
- SA supplemented water Abbreviated as SA supplemented water
- Example 11 Expanded cultured cells cryopreserved in (1) and the same donor prepared in the same manner as in Example 1— (1) PBMC were rapidly thawed in a 37 ° C. water bath. Peripheral blood lymphocyte concentration (hereinafter referred to as PBL) concentration was calculated assuming that the CD3-positive cell ratio in PBMC was 70%.
- PBL Peripheral blood lymphocyte concentration
- As a solvent use 4% HS A saline solution, and use 0.5 X 10 7 cells / mouse with a part of PBL as feeder, and 1.0 x 10 8 cells / mouse after PBL and expanded culture. Mice were suspended in a solvent to obtain the required number of cells, and used as cells for administration. Prior to cell administration, all mice were irradiated with X-rays (0.090 CZkg), and the prepared cells were administered intraperitoneally in the order of group A force in the order of 0.3 mL.
- the H-HL-2 used during T-cell expansion culture was prepared with 0.4% HSA-enriched diet so that it became 2 X 10 4 UZ mice, and administration of the cells was completed in Example 11- (4) Mouse force 0.2 mL was administered intraperitoneally. Human IL-2 was administered four times, once a day from the day of cell administration.
- Body weight was measured at appropriate intervals from the administration day to the 21st day. As a result, in group B that received PBL, the body weight decreased on the 8th day, and one died on the 13th day. In the other groups, all died without dying until day 21. Looking at the rate of weight loss, in Group A and Group C, there was a change, but on the 21st day the initial weight was maintained, and in Group D there was a slight decrease. In the Xeno-GVHD response with weight loss as an indicator, PBL showed a dominant response, indicating that expanded cells in CH-296 fixed condition were more effective than those in non-fixed condition .
- the spleen was removed at the time of necropsy on the 21st day after the start of cell administration, and the ratio of human CD3-positive cells in the spleen was analyzed by flow cytometry.
- group B one mouse died on the 13th day, so the mouse removed the spleen immediately after death, and the other mouse was necropsied on the 13th day and the spleen was removed.
- the excised spleen was ground with a slide glass, filtered with nylon mesh, and centrifuged. The supernatant was removed, hemolyzed with ACK buffer, and washed with RPMI 1640 medium to prepare cells.
- FITC-labeled mouse anti-human CD3 antibody (Dako) was used.
- the CD3 positive rate in splenocytes in group B analyzed on day 13 was approximately 70%, and 21 It was about 59% in group D analyzed on the day.
- the positive rate was less than 1% in group A and several percent in group C. From this, it was found that the expanded cultured cells under CH-296 fixed condition had a higher survival rate in the spleen compared to the non-fixed condition and were maintained for 21 days.
- Example 12 Expansion of lymphocytes supplemented with IL-2, IL12, IFN- ⁇ , and anti-IL-4 antibodies
- Example 1 Performed in the same manner as (2). However, ACD-A solution ( ⁇ 5.0) was used to immobilize the anti-human CD3 antibody and CH-296 fragment, and the amount of ACD- ⁇ solution added to the 12-well cell culture plate was changed to 0.45 mL.
- Example 12 Anti-human prepared in Example 12— (1) after suspending PBMC prepared in Example 1— (1) to 0.5% GT—T503 at 0.25 ⁇ 10 6 cells / mL Add 0.5% GT-T503 to the CD3 antibody-fixed antibody plate or anti-human CD3 antibody and CH-296 immobilized plate with 0.5 mL LZ well, and add 1 mL of cell solution to each cell solution. The plates were cultivated and cultured at 37 ° C in 5% CO (culture day 0). On day 4 of culture start, add 0.5% of the culture solution for each group.
- IL-12 (R & D Systems) 50UZmL, IFN- ⁇ (R & D Systems) 20ngZmL, anti-HIML-4 antibody (Betaton Dickinson) (Manufactured)
- the final concentration of 2 gZmL was added, and on the 7th and 11th days of culture, each cyto-in antibody was added to the newly added medium so as to achieve the above final concentration.
- the number of viable cells was counted by trypan blue staining, and the expansion culture rate was calculated by comparison with the number of cells at the start of culture. Result The results are shown in Table 31.
- Example 12 Cells prepared in (2) were stained with each antibody in the same manner as in Example 1— (4) and analyzed. However, antibody combinations were performed as follows. Specifically, as a negative control, FITC-labeled mouse IgGlZRDl-labeled mouse IgGlZPC5-labeled mouse IgGl + ECD-labeled mouse IgGl, RD1-labeled mouse anti-human CD45RA antibody ZF ITC-labeled mouse anti-human CCR7 antibody ZECD-labeled mouse anti-human CD4 antibody ZPC5-labeled mouse anti-human CD8 Antibody or RD1-labeled mouse anti-human CD45RA antibody ZFITC-labeled mouse anti-human CD62L antibody ZECD-labeled mouse anti-human CD4 antibody ZPC5-labeled mouse anti-human CD8 antibody was used for staining.
- the cells were subjected to flow cytometry, and the ratio of CD45RA + CCR7 + T cells and CD45RA + CD62L + T cells in the entire T cell region, CD8 + T cell region or CD4 + T cell region was calculated. The results are shown in Table 32.
- Table 32 in the group using the culture vessel in which the CH-296 fragment was fixed, the ⁇ during culture was compared with the control group. CD45RA + CCR7 + T cells in cells The population and CD45RA + CD62L + T cell population gave high results. From the above, in cultures in which IL 2, IL 12, IFN- ⁇ , and anti-human IL-4 antibodies were added to GT T503 medium, the CD45R of sputum cells can be obtained by coexisting the CH-296 fragment in the early stage of expansion culture. It was revealed that the sputum cell population can be expanded while increasing the A + CCR7 + or CD45RA + CD62L + T cell population ratio.
- Example 13 Freshly isolated PBMC Kachin et al. CH-296 Allogeneic MLR of cell population expanded using gas permeable culture bag after initial stimulation
- PBMC was separated from the collected blood in the same manner as in Example 1- (1).
- the number of cells of the collected PBMC was calculated by Triban Blue and Turku staining method (Turku Solution, Nakarai Tester), and used for each experiment without being cryopreserved.
- Example 6 Performed in the same manner as in (1).
- Example 13- (2) Suspend the PBMC prepared in Example 13- (1) to 0.5 ⁇ 10 6 cellsZmL in 5% autologous plasma GT—T503, prepare the cell solution, and then prepare in Example 13- (2). 0.5% GT-T503 was added to the anti-human CD3 antibody-immobilized flask or anti-human CD3 antibody and CH-296-immobilized flask, and the cell solution was added to each 9 mLZ flask. IL-2 was added to a final concentration of lOOOUZmL, and these flasks were cultured at 37 ° C in 5% CO (culture day 0). On day 4 of culture, 21 mL of each group of culture broth
- CH- 296 group 6
- CH- 296 group 6
- Example 13 Cells prepared in (3) were stained with each antibody and analyzed in the same manner as in Example 6— (3). The results are shown in Table 34.
- CD 28 + CD 2 7 Spider cells 6 5. 54% 7 6. 42%
- the CH-296 group showed a high value for any cell surface marker as compared to the control group.
- Example 13 Cells on the 14th day of culture prepared in (3) were cryopreserved and thawed in the same manner as in Example 5- (4) and used for each experiment.
- Allogeneic MLR was performed in the same manner as in Example 6 (4) using the cells prepared in (5). However, the following points were changed. Stimulator cell and Responder cell concentration adjusted to 1 or 2 X 10 6 cells / mL, and after suspending cells in each tool on the 5th day of culture, halve each into 2 wells , 5HRPMI 1mL containing 100 OUZmL IL-2 was added to each well, and the culture was continued until day 7.
- Example 13 On the 7th day after the start of culture, the number of viable cells was counted by trypan blue staining on the cells obtained in (6), and the expansion culture rate was calculated by comparison with the number of cells at the start of culture. The results are shown in Table 35.
- Example 13 The cytotoxic activity of the cells on day 7 after the start of induction prepared in (6) was carried out in the same manner as in Example 6-(6). However, autologous PBMC or non-self PBMC blasted with PHA for 7 days as target cells at this time were used as force lusein-labeled target cells. When measuring cytotoxic activity, 30 times the amount of K562 cells was mixed with force lucein-labeled target cells. The EZT ratio was set to 90-3. The results of the cytotoxic activity measurement are shown in Table 36.
- allogeneic MLR provides a cell population with a stronger alloantigen-specific cytotoxic activity in cultured cells using CH-296 fixed in the initial stage of T cell population expansion culture. It became.
- Example 14 Induction of Allogeneic MLR and anti-MART-1 CTL in expanded cell population using gas-permeable culture bag during initial stimulation of CH-296 and subsequent culture
- Anti-human CD3 antibody and CH-296 were immobilized on the culture equipment used in the following experiments. That is, a gas permeable culture bag (culture area 75 cm 2 , manufactured by Takara Bio Co., Ltd., product code
- KB620 was supplemented with 9 mL of PBS containing anti-human CD3 antibody (final concentration 5 ⁇ g / mL). At this time, the same amount of PBS containing anti-human CD3 antibody (final concentration 5 / z gZmL) and CH-296 final concentration (25 ⁇ g / mL) was added to the CH-296 addition group.
- Example 13- (3) The same procedure as in Example 13- (3) was used except that the bag prepared in Example 14- (1) was used instead of the culture flask as the culture equipment during CH-296 stimulation. After starting culture
- CH-296 As shown in Table 37, during initial stimulation with CH-296 and subsequent culture using a gas-permeable culture bag, CH-296 was fixed in the early stage of expanded cell population expansion. In the group using the culture equipment (hereinafter referred to as CH-296 group), the expansion rate of the sputum cell population was higher than that in the control group.
- Example 14 Cells prepared in (2) were stained with each antibody and analyzed in the same manner as in Example 6— (3). The results are shown in Table 38.
- the CH-296 group showed a high value for any cell surface marker as compared to the control group.
- Example 14 The cytotoxic activity of the cells 10 days after the start of induction prepared in (1) was performed in the same manner as in Example 6- (6), except that an EZT ratio of 90 was newly set. . Table 39 shows the results of the measurement of cytotoxic activity.
- Non-self P I I A 9 0 6 5. 0 5 7 2. 7 3 Blast cells 3 0 4 4. 9 2 6 1. 4 3
- Example 14 Cultured day 14 cells prepared in (2) were cryopreserved and thawed in the same manner as in Example 5— (4) and used for each experiment.
- Example 1- (1) and 14- (6) induction of anti-tumor-associated antigen (MART-1) -specific CTL was performed in the same manner as in Example 5- (5). The culture was continued for 14 days. However, the following points were changed. Responder cells are suspended in 5HRPMI at 1 X 10 6 cells / mL, and 0.5 mL Zwell is added to each 24-well cell culture plate. On the third day after the start of culture, half of the culture supernatant was removed, 5H RPMI containing 1mL of 60U / mL IL-2 was added to each well, and after 1 week, the cultured cells were added to 1.5-3. the OX10 6 cells ZmL, including antigen-presenting cells was adjusted to 1.
- OX10 6 cellsZmL that was added Caro by 0.5MLZwell, after removing the culture supernatant half after 10 days of culture, the IL- 2 of 60U / mL 5HR PMI lmL was added to each well.
- Example 14-1 Measurement of the cytotoxic activity of CTL on the 14th day after the start of induction prepared in Example 14-1 (7) was performed in the same manner as in Example 6- (9). The results of the cytotoxic activity measurement are shown in Table 40.
- Example 15 Cells derived from CD45RA + CCR7 + CD8 + T cells and CD45RA-CCR7-CD8 + T cells derived from expanded cells using gas-permeable culture bags during initial stimulation of CH-296 and subsequent culture Anti-MART— 1CTL induction
- Example 1 (1) and Example 14 (6) were separated into CD45RA + CCR7 + CD8 + and CD45RA + CCR7-CD8 + fractions in the same manner as in Example 7- (1). Each was isolated to obtain a fraction having a purity of 93% to 99%.
- Example 7- (1) The cells prepared in Example 1 (1) and Example 14 (6) were separated into CD45RA + CCR7 + CD8 + and CD45RA + CCR7-CD8 + fractions in the same manner as in Example 7- (1). Each was isolated to obtain a fraction having a purity of 93% to 99%.
- MART-1 anti-tumor-associated antigen
- Example 15 Using the cells prepared in (1), induction of anti-tumor-associated antigen (MART-1) -specific CTL was carried out in the same manner as in Example 14— (7), and the culture was continued for 14 days. However, the following points were changed. Responder cells were suspended in 5 HRPMI to 1 X 10 6 cells ZmL, and 0.25 mL Zwell was added to each 48-well cell culture plate. On the first day of culture, 5 HRPMI0 containing 60 U / mL IL-2 5 mL was added to each well, half of the culture supernatant was removed on the 3rd day of culture, and 60 U / mL IL-2 was contained. 5 HRPMIO. 5 mL was added to each well, and 1 week.
- MART-1 anti-tumor-associated antigen
- culture cells were adjusted to 0.2 to 1.7 X 10 6 cellsZmL, antigen-presenting cells were adjusted to 1.OX 10 6 cellsZmL, and 0.5 mLZwell was added to each 24-well cell culture plate. On the 10th day after starting, half of the culture supernatant was removed, and then 5 mL of 5HRPMI containing 60 U / mL IL-2 was added to each well.
- Example 15 On the 14th day after the start of culture, the number of viable cells was measured by trypan blue staining for the cells obtained in (2), and the expanded culture rate was calculated by comparison with the number of cells at the start of culture. It was. The results are shown in Table 41.
- CD45RA + CCR7 + CD 8 isolated from cultured cells (hereinafter referred to as CH-296 group) using culture equipment with CH-296 fixed in the early stage of T cell population expansion culture.
- CH-296 group the expanded culture rate of the CTL population was higher than that in the control group.
- a larger CTL population can be obtained by inducing CTL from CD45RA + CCR7 + T cells contained in a high proportion of cultured cells using CH-296 fixed in the early stage of T cell population expansion culture. It became clear that it was obtained.
- Example 15 The cytotoxic activity of CTL 14 days after the start of induction prepared in (2) was the same as in Example 6- (9) except that the E / T ratio was set to 30-3. went. The results of the cytotoxic activity measurement are shown in Table 42.
- Example 14- (7) anti-tumor-associated antigen (MART-1) -specific CTL was induced in the same manner as in Example 14- (7). The culture was continued for days.
- MART-1 anti-tumor-associated antigen
- Example 16 The antigen recognition ability of CTL 15 days after initiation of induction prepared in (1) was -Measured in the same manner as (2). Table 43 shows the results of antigen recognition ability measurement. The antigen recognition ability was expressed as the relative value of the cytotoxic activity at each additional peptide concentration when the cytotoxic activity when the peptide concentration added to the target cell was 10 ⁇ 10 was taken as 100.
- Example 17 Evaluation of cytoforce-in productivity of cell-derived CD45RA + CCR7 + T cells expanded using a gas-permeable culture bag during initial stimulation and subsequent culture of CH-296
- Example 14 Cells prepared in one (6) were stained and sorted in the same manner as in Example 3- (3).
- Example 17 Suspend each cell population obtained in (1) in 0.5% GT-T503, and add cells to each panel to 2 ⁇ 10 5 cells / 0.2 mL on the prepared plate. And cultured at 37 ° C in 5% CO. 24 hours later
- CD45RA + T cells and CD8-T cells were dominant in IL-2 production in CD45RA + CCR7 + T cells.
- the CD45RA + CCR 7+ T cell population of frozen cells bag-cultured with CH-296 was used as naive T-like cells. It was shown to have the nature of
- Example 4 Performed in the same manner as in (1). However, the CH-296 fragment was not used. Instead, the H-296, CH-271, H-271, and C-CS1 fragments at the final concentration (25 ⁇ g / mL) were used for the fragment-added column. It added so that it might become.
- Example 18 The proportion of CD45RA + CCR7 + T cells on day 9 of culture was calculated for the cells prepared in (2) in the same manner as in Example 3— (2). The results are shown in Table 46.
- the CD45RA + CCR7 + T cell population was higher in the T cell population in culture than in the control group.
- the present invention provides a method for producing a T cell population.
- This method is suitable for immunotherapy, for example, as a T cell population that contains a high ratio of T cells that express CD45RA and express at least one selected from the group consisting of CD62L, CCR7, CD27, and CD28. used. Therefore, the method of the present invention is expected to make a great contribution to the medical field.
- Fig. 1 is a schematic diagram showing the domain structure of fibronectin. [2] It is a figure showing the tumor formation inhibitory effect of mice by T cell administration. Sequence listing free text
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Cell Biology (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- Mycology (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Oncology (AREA)
- General Engineering & Computer Science (AREA)
- Virology (AREA)
- Biochemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Hematology (AREA)
- Communicable Diseases (AREA)
- Molecular Biology (AREA)
- Gastroenterology & Hepatology (AREA)
- Physics & Mathematics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Plant Pathology (AREA)
- Biophysics (AREA)
- Pulmonology (AREA)
- Developmental Biology & Embryology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
Claims
Priority Applications (9)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2007538714A JP5156382B2 (ja) | 2005-09-30 | 2006-09-27 | T細胞集団の製造方法 |
EP06810596A EP1939278A4 (en) | 2005-09-30 | 2006-09-27 | METHOD FOR PRODUCING A T CELL POPULATION |
AU2006298188A AU2006298188B2 (en) | 2005-09-30 | 2006-09-27 | Method for production of T cell population |
US11/992,661 US20100068192A1 (en) | 2005-09-30 | 2006-09-27 | Method for Production of T Cell Population |
EA200800996A EA016168B1 (ru) | 2005-09-30 | 2006-09-27 | Способ получения т-клеточной популяции и ее применение |
CA002623735A CA2623735A1 (en) | 2005-09-30 | 2006-09-27 | Method for production of t cell population |
CN2006800441169A CN101400785B (zh) | 2005-09-30 | 2006-09-27 | T细胞群的制备方法 |
KR1020087010395A KR101408565B1 (ko) | 2005-09-30 | 2006-09-27 | T 세포 집단의 제조 방법 |
HK09108701.4A HK1130509A1 (en) | 2005-09-30 | 2009-09-23 | Method for production of t cell population t |
Applications Claiming Priority (8)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2005288983 | 2005-09-30 | ||
JP2005-288983 | 2005-09-30 | ||
JP2006-102103 | 2006-04-03 | ||
JP2006102103 | 2006-04-03 | ||
JP2006-196950 | 2006-07-19 | ||
JP2006196950 | 2006-07-19 | ||
JP2006241773 | 2006-09-06 | ||
JP2006-241773 | 2006-09-06 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2007040105A1 true WO2007040105A1 (ja) | 2007-04-12 |
Family
ID=37906148
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2006/319105 WO2007040105A1 (ja) | 2005-09-30 | 2006-09-27 | T細胞集団の製造方法 |
Country Status (11)
Country | Link |
---|---|
US (1) | US20100068192A1 (ja) |
EP (1) | EP1939278A4 (ja) |
JP (1) | JP5156382B2 (ja) |
KR (1) | KR101408565B1 (ja) |
CN (1) | CN101400785B (ja) |
AU (1) | AU2006298188B2 (ja) |
CA (1) | CA2623735A1 (ja) |
EA (1) | EA016168B1 (ja) |
HK (1) | HK1130509A1 (ja) |
TW (1) | TW200741001A (ja) |
WO (1) | WO2007040105A1 (ja) |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007142300A1 (ja) * | 2006-06-09 | 2007-12-13 | Takara Bio Inc. | リンパ球の製造方法 |
WO2008143255A1 (ja) * | 2007-05-22 | 2008-11-27 | Takara Bio Inc. | 細胞集団の製造方法 |
WO2008143014A1 (ja) * | 2007-05-11 | 2008-11-27 | Takara Bio Inc. | がん治療剤 |
JP2010527606A (ja) * | 2007-05-23 | 2010-08-19 | サンガモ バイオサイエンシーズ, インコーポレイテッド | 導入遺伝子の発現を増強するための方法および組成物 |
WO2020032179A1 (ja) * | 2018-08-10 | 2020-02-13 | 国立大学法人京都大学 | Cd3陽性細胞の製造方法 |
Families Citing this family (16)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP5485139B2 (ja) * | 2008-03-27 | 2014-05-07 | タカラバイオ株式会社 | 遺伝子導入細胞の製造方法 |
CN101706507B (zh) * | 2009-07-27 | 2013-05-01 | 浙江大学 | 用异硫氰酸荧光素标记的抗人cd45ra单抗的应用 |
CN101831434B (zh) * | 2009-07-27 | 2012-05-09 | 浙江大学 | 抗人cd45ra鼠免疫球蛋白可变区基因及用途 |
US9062287B2 (en) | 2009-09-11 | 2015-06-23 | Takara Bio Inc. | Process for production of natural killer cells |
WO2013126712A1 (en) | 2012-02-22 | 2013-08-29 | The Trustees Of The University Of Pennsylvania | Compositions and methods for generating a persisting population of t cells useful for the treatment of cancer |
CN108498532B (zh) | 2012-05-09 | 2021-07-23 | 坎泰克斯制药股份有限公司 | 骨髓抑制的治疗 |
DK2981607T3 (da) * | 2013-04-03 | 2020-11-16 | Memorial Sloan Kettering Cancer Center | Effektiv generering af tumormålrettede t-celler afledt af pluripotente stamceller |
WO2016133907A1 (en) * | 2015-02-17 | 2016-08-25 | Cantex Pharmaceuticals, Inc. | Adoptive cell transfer methods |
WO2016133910A1 (en) | 2015-02-17 | 2016-08-25 | Cantex Pharmaceuticals, Inc. | Treatment of cancers and hematopoietic stem cell disorders privileged by cxcl12-cxcr4 interaction |
EP3091032A1 (en) * | 2015-05-08 | 2016-11-09 | Miltenyi Biotec GmbH | Humanized antibody or fragment thereof specific for cd3 |
MA45488A (fr) * | 2015-10-22 | 2018-08-29 | Juno Therapeutics Gmbh | Procédés, kits et appareil de culture de cellules |
ES2957890T3 (es) | 2016-04-08 | 2024-01-29 | Univ Emory | Métodos de tratamiento del cáncer y las enfermedades infecciosas mediante terapias celulares |
WO2017214939A1 (zh) * | 2016-06-16 | 2017-12-21 | 毛侃琅 | 提升 ccr7 基因表达水平的慢病毒表达载体及其应用 |
SG11202102654UA (en) * | 2018-09-19 | 2021-04-29 | Fujifilm Cellular Dynamics Inc | Protein l for activation and expansion of chimeric antigen receptor-modified immune cells |
WO2020102701A1 (en) * | 2018-11-16 | 2020-05-22 | Rapa Therapeutics, Llc | Methods for the manufacture of th1/tc1 phenotype t cells |
CN118786208A (zh) * | 2022-02-16 | 2024-10-15 | 加拿大干细胞技术公司 | 用于扩增淋巴细胞的组合物和方法 |
Citations (10)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5102988A (en) | 1988-06-30 | 1992-04-07 | Takara Shuzo Co., Ltd. | Polypeptide with cell-spreading activity |
US5198423A (en) | 1989-05-26 | 1993-03-30 | Takara Shuzo Co., Ltd. | Functional polypeptide containing a cell binding domain and a heparin binding domain of fibronectin |
WO1997018318A1 (en) | 1995-11-13 | 1997-05-22 | Takara Shuzo Co., Ltd. | Method for gene introduction into target cells by retrovirus |
JP2729712B2 (ja) | 1991-04-23 | 1998-03-18 | 寳酒造株式会社 | 機能性ポリペプチド |
WO2000009168A1 (en) | 1998-08-11 | 2000-02-24 | The United States Of America, Represented By Department Of Health And Human Services | A method of transducing mammalian cells, and products related thereto |
JP3104178B2 (ja) | 1990-03-30 | 2000-10-30 | 寶酒造株式会社 | 機能性ポリペプチド |
WO2003016511A1 (en) | 2001-08-15 | 2003-02-27 | Takara Bio Inc. | Method of extended culture for antigen-specific cytotoxic t lumphocytes |
WO2003080817A1 (en) | 2002-03-25 | 2003-10-02 | Takara Bio Inc. | Process for producing cytotoxic lymphocyte |
WO2004018667A1 (ja) * | 2002-08-26 | 2004-03-04 | Kirin Beer Kabushiki Kaisha | ペプチド及びこれを含む医薬 |
WO2005019450A1 (ja) | 2003-08-22 | 2005-03-03 | Takara Bio Inc. | 細胞傷害性リンパ球の製造方法 |
Family Cites Families (16)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5188959A (en) * | 1989-09-28 | 1993-02-23 | Trustees Of Tufts College | Extracellular matrix protein adherent t cells |
US6821778B1 (en) * | 1993-12-01 | 2004-11-23 | The Board Of Trustees Of Leland Stanford Junior University | Methods for using dendritic cells to activate gamma/delta-T cell receptor-positive T cells |
DE4412794A1 (de) * | 1994-04-14 | 1995-12-14 | Univ Ludwigs Albert | Verfahren zur Herstellung von dendritischen Zellen, so erhaltene Zellen und Behälter zur Durchführung dieses Verfahrens |
US5827642A (en) * | 1994-08-31 | 1998-10-27 | Fred Hutchinson Cancer Research Center | Rapid expansion method ("REM") for in vitro propagation of T lymphocytes |
US7067318B2 (en) * | 1995-06-07 | 2006-06-27 | The Regents Of The University Of Michigan | Methods for transfecting T cells |
US6692964B1 (en) * | 1995-05-04 | 2004-02-17 | The United States Of America As Represented By The Secretary Of The Navy | Methods for transfecting T cells |
EP0824594B1 (en) * | 1995-05-04 | 2005-04-06 | The United States of America as Representend by The Secretary of the Navy | Improved methods for transfecting t cells |
US6734014B1 (en) * | 1996-02-08 | 2004-05-11 | The United States Of America As Represented By The Department Of Health And Human Services | Methods and compositions for transforming dendritic cells and activating T cells |
US6316257B1 (en) * | 1996-03-04 | 2001-11-13 | Targeted Genetics Corporation | Modified rapid expansion methods (“modified-REM”) for in vitro propagation of T lymphocytes |
TWI239352B (en) * | 1997-07-23 | 2005-09-11 | Takara Bio Inc | Gene transfer method with the use of serum-free medium |
KR100674140B1 (ko) * | 1999-03-23 | 2007-01-26 | 다카라 바이오 가부시키가이샤 | 유전자 치료제 |
KR20020010206A (ko) * | 2000-07-27 | 2002-02-04 | 이시우 | 인터루킨 12와 보조활성인자 b7.1 유전자를 함유하는dna 벡터 및 이벡터가 도입된 항암 세포백신 |
US7816134B2 (en) * | 2000-08-16 | 2010-10-19 | Takara Bio Inc. | Method of extensive culture of antigen-specific cytotoxic T cells |
EP1401495A4 (en) * | 2001-06-01 | 2005-11-23 | Xcyte Therapies Inc | T CELL-INDUCED TISSUE PARA- TURE AND REGENERATION |
US7745140B2 (en) * | 2002-01-03 | 2010-06-29 | The Trustees Of The University Of Pennsylvania | Activation and expansion of T-cells using an engineered multivalent signaling platform as a research tool |
US20080227504A1 (en) * | 2007-03-15 | 2008-09-18 | Shih-Yang Chan | Ceramic casing device for mobile handset |
-
2006
- 2006-09-27 CA CA002623735A patent/CA2623735A1/en not_active Abandoned
- 2006-09-27 WO PCT/JP2006/319105 patent/WO2007040105A1/ja active Application Filing
- 2006-09-27 AU AU2006298188A patent/AU2006298188B2/en not_active Ceased
- 2006-09-27 CN CN2006800441169A patent/CN101400785B/zh not_active Expired - Fee Related
- 2006-09-27 US US11/992,661 patent/US20100068192A1/en not_active Abandoned
- 2006-09-27 JP JP2007538714A patent/JP5156382B2/ja not_active Expired - Fee Related
- 2006-09-27 EP EP06810596A patent/EP1939278A4/en not_active Withdrawn
- 2006-09-27 EA EA200800996A patent/EA016168B1/ru not_active IP Right Cessation
- 2006-09-27 KR KR1020087010395A patent/KR101408565B1/ko not_active IP Right Cessation
- 2006-09-28 TW TW095136081A patent/TW200741001A/zh unknown
-
2009
- 2009-09-23 HK HK09108701.4A patent/HK1130509A1/xx not_active IP Right Cessation
Patent Citations (10)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5102988A (en) | 1988-06-30 | 1992-04-07 | Takara Shuzo Co., Ltd. | Polypeptide with cell-spreading activity |
US5198423A (en) | 1989-05-26 | 1993-03-30 | Takara Shuzo Co., Ltd. | Functional polypeptide containing a cell binding domain and a heparin binding domain of fibronectin |
JP3104178B2 (ja) | 1990-03-30 | 2000-10-30 | 寶酒造株式会社 | 機能性ポリペプチド |
JP2729712B2 (ja) | 1991-04-23 | 1998-03-18 | 寳酒造株式会社 | 機能性ポリペプチド |
WO1997018318A1 (en) | 1995-11-13 | 1997-05-22 | Takara Shuzo Co., Ltd. | Method for gene introduction into target cells by retrovirus |
WO2000009168A1 (en) | 1998-08-11 | 2000-02-24 | The United States Of America, Represented By Department Of Health And Human Services | A method of transducing mammalian cells, and products related thereto |
WO2003016511A1 (en) | 2001-08-15 | 2003-02-27 | Takara Bio Inc. | Method of extended culture for antigen-specific cytotoxic t lumphocytes |
WO2003080817A1 (en) | 2002-03-25 | 2003-10-02 | Takara Bio Inc. | Process for producing cytotoxic lymphocyte |
WO2004018667A1 (ja) * | 2002-08-26 | 2004-03-04 | Kirin Beer Kabushiki Kaisha | ペプチド及びこれを含む医薬 |
WO2005019450A1 (ja) | 2003-08-22 | 2005-03-03 | Takara Bio Inc. | 細胞傷害性リンパ球の製造方法 |
Non-Patent Citations (32)
Title |
---|
BENIGNI F, J. IMMUNOL., vol. 175, no. 2, 2005, pages 739 - 748 |
DARDALHON V. ET AL.: "Highly efficient gene transfer in naive human T cells with a murine leukemia virus-based vector", BLOOD, vol. 96, no. 3, 2000, pages 885 - 893, XP003011564 * |
DAVIS L.S. ET AL.: "Fibronectin promotes proliferation of naive and memory T cells by signaling through both the VLA-4 and VLA-5 integrin molecules", J. IMMUNOL., vol. 145, no. 3, 1990, pages 785 - 793, XP002381603 * |
ESCHERICHIA COLI, 12 May 1989 (1989-05-12) |
ESCHERICHIA COLI, 17 June 1988 (1988-06-17) |
ESCHERICHIA COLI, 30 January 1989 (1989-01-30) |
ESCHERICHIA COLI, 5 March 1990 (1990-03-05) |
ESCHERICHIA COLI, 8 April 1991 (1991-04-08) |
ESCHERICHIA COLI, August 1991 (1991-08-01) |
GATTINONI L. ET AL.: "Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells", J. CLIN. INVEST., vol. 115, no. 6, June 2005 (2005-06-01), pages 1616 - 1626, XP003011560 * |
GATTINONI L., J. CLIN. INVEST., vol. 115, no. 6, 2005, pages 1616 - 1626 |
HALVORSON M.J. ET AL.: "alpha4 and alpha5 integrins costimulate the CD3-dependent proliferation of fetal thymocytes", CELL IMMUNOL., vol. 189, no. 1, 1998, pages 1 - 9, XP003002658 * |
HANENBERG H. FIVE, HUMAN GENE THERAPY, vol. 8, no. 18, 1997, pages 2193 - 2206 |
IDENO M. ET AL.: "RetroNectin o Kumiawaseta T Saibo Kakudai Baiyo (II): Kakudai Baiyo sareta T Saibo Shudan wa NaiveT-yo Saibo no Hiritsu ga Takaku, Takai Kogen Ninshikino o Hakki", DAI 65 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION KIJI, 28 August 2006 (2006-08-28), pages 330, P-708, XP003011559 * |
IDENO M. ET AL.: "Soshikitai Human Fibronectin Fragment Shigeki ni yoru Kasseika CTL no Tairyo Expansion-ho", DAI 62 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION KIJI, 2003, pages 175, 2144-OA, XP003011567 * |
KIMIDUKA F. ET AL., BIOCHEM., vol. 110, 1991, pages 284 - 291 |
KIMIZUKA F., J. BIOCHEM., vol. 110, no. 2, 1991, pages 284 - 291 |
KORNBRIHTT A. R. ET AL., EMBO J., vol. 4, no. 7, 1985, pages 1755 - 1759 |
LICHTENFELS R. ET AL., J. IMMUNOL., vol. 172, no. 2, 1994, pages 227 - 239 |
MATSUYAMA T. ET AL.: "Activation of CD4 cells by fibronectin and anti-CD3 antibody. A synergistic effect mediated by the VLA-5 fibronectin receptor complex", J. EXP. MED., vol. 170, no. 4, 1989, pages 1133 - 1148, XP003011562 * |
MURAKI N. ET AL.: "RetroNectin o Kumiawaseta T Saibo Kakudai Baiyo (I): Kakudai Baiyo ga Anteika shi, NaiveT-yo Saibo ga Kohiritsu de Zoshoku (T cell expansion using RetroNectin (I): Useful method to expand Tcells, characterized by high portion of NaiveT-like cells)", DAI 65 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION KIJI, 28 August 2006 (2006-08-28), pages 330, P-707, XP003011558 * |
PLASMID ENCODING, 23 July 2004 (2004-07-23) |
PLEBANSKI M. ET AL., EUR. J. IMMUNOL, vol. 25, no. 6, 1995, pages 1783 - 1787 |
RIDDELL S.R. ET AL.: "The use of anti-CD3 and anti-CD28 monolocnal antibodies to clone and expand human antigen-specific T cells", J. IMMUNOL. METHODS, vol. 128, no. 2, 1990, pages 189 - 201, XP003011565 * |
RUOSLAHTI E. ET AL., J. BIOL. CHEM., vol. 256, no. 14, 1981, pages 7277 - 7281 |
SAGAWA H. ET AL.: "Ko-CD3 Kotai no RetroNectin o Kumiawaseru Koto ni yoru LAK Saibo Inyu Ryoho no Kairyo", DAI 62 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION KIJI, 2003, pages 438, 2624-PP, XP003011566 * |
See also references of EP1939278A4 |
SEKIGUCHI K. ET AL., BIOCHEMISTRY, vol. 25, no. 17, 1986, pages 4936 - 4941 |
SIMON M.M. ET AL.: "The outer surface lipoprotein A of Borrelia burgdorferi provides direct and indirect augmenting/co-stimulatory signals for the activation of CD4+ and CD8+ T cells", IMMUNOL. LETT., vol. 46, no. 3, 1995, pages 137 - 142, XP003011563 * |
STURM A. ET AL.: "Dual function of the extracellular matrix: stimulatory for cell cycle progression of naive T cells and antiapoptotic for tissue-derived memory T cells", J. IMMUNOL., vol. 173, no. 6, 2004, pages 3889 - 3900, XP003011561 * |
VALMORI D. ET AL., J. IMMUNOL., vol. 160, 1998, pages 1750 - 1758 |
WILLIAMS D. A. ET AL., NATURE, vol. 352, 1991, pages 438 - 441 |
Cited By (10)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007142300A1 (ja) * | 2006-06-09 | 2007-12-13 | Takara Bio Inc. | リンパ球の製造方法 |
US8216837B2 (en) | 2006-06-09 | 2012-07-10 | Takara Bio Inc. | Method of producing lymphocytes |
WO2008143014A1 (ja) * | 2007-05-11 | 2008-11-27 | Takara Bio Inc. | がん治療剤 |
JPWO2008143014A1 (ja) * | 2007-05-11 | 2010-08-05 | タカラバイオ株式会社 | がん治療剤 |
WO2008143255A1 (ja) * | 2007-05-22 | 2008-11-27 | Takara Bio Inc. | 細胞集団の製造方法 |
JPWO2008143255A1 (ja) * | 2007-05-22 | 2010-08-12 | タカラバイオ株式会社 | 細胞集団の製造方法 |
JP2010527606A (ja) * | 2007-05-23 | 2010-08-19 | サンガモ バイオサイエンシーズ, インコーポレイテッド | 導入遺伝子の発現を増強するための方法および組成物 |
WO2020032179A1 (ja) * | 2018-08-10 | 2020-02-13 | 国立大学法人京都大学 | Cd3陽性細胞の製造方法 |
JPWO2020032179A1 (ja) * | 2018-08-10 | 2021-08-26 | 国立大学法人京都大学 | Cd3陽性細胞の製造方法 |
JP7382603B2 (ja) | 2018-08-10 | 2023-11-17 | 国立大学法人京都大学 | Cd3陽性細胞の製造方法 |
Also Published As
Publication number | Publication date |
---|---|
JPWO2007040105A1 (ja) | 2009-04-16 |
JP5156382B2 (ja) | 2013-03-06 |
HK1130509A1 (en) | 2009-12-31 |
EP1939278A4 (en) | 2009-06-03 |
US20100068192A1 (en) | 2010-03-18 |
AU2006298188B2 (en) | 2012-03-15 |
EA016168B1 (ru) | 2012-02-28 |
EP1939278A1 (en) | 2008-07-02 |
CA2623735A1 (en) | 2007-04-12 |
KR101408565B1 (ko) | 2014-06-17 |
EA200800996A1 (ru) | 2008-08-29 |
KR20080056266A (ko) | 2008-06-20 |
CN101400785B (zh) | 2013-09-25 |
TW200741001A (en) | 2007-11-01 |
CN101400785A (zh) | 2009-04-01 |
AU2006298188A1 (en) | 2007-04-12 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP5156382B2 (ja) | T細胞集団の製造方法 | |
US8975070B2 (en) | Process for producing cytotoxic lymphocyte | |
US8765469B2 (en) | Method of producing lymphocytes | |
JP5805089B2 (ja) | 細胞集団の製造方法 | |
WO2011024791A1 (ja) | レチノイン酸存在下でのt細胞集団の製造方法 | |
JPWO2007142300A1 (ja) | リンパ球の製造方法 | |
JP4741906B2 (ja) | リンパ球の製造方法 | |
JP5485139B2 (ja) | 遺伝子導入細胞の製造方法 | |
JPWO2008143255A1 (ja) | 細胞集団の製造方法 | |
JP2010099022A (ja) | リンパ球の製造方法 | |
JP2010063455A (ja) | リンパ球の製造方法 | |
JP2010094123A (ja) | リンパ球の製造方法 | |
MX2008004225A (es) | Metodo para la produccion de una poblacion de celulas t |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
WWE | Wipo information: entry into national phase |
Ref document number: 200680044116.9 Country of ref document: CN |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
ENP | Entry into the national phase |
Ref document number: 2007538714 Country of ref document: JP Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 2623735 Country of ref document: CA |
|
WWE | Wipo information: entry into national phase |
Ref document number: 11992661 Country of ref document: US |
|
WWE | Wipo information: entry into national phase |
Ref document number: MX/a/2008/004225 Country of ref document: MX Ref document number: 2006810596 Country of ref document: EP |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2006298188 Country of ref document: AU |
|
WWE | Wipo information: entry into national phase |
Ref document number: 1020087010395 Country of ref document: KR Ref document number: 200800996 Country of ref document: EA |
|
ENP | Entry into the national phase |
Ref document number: 2006298188 Country of ref document: AU Date of ref document: 20060927 Kind code of ref document: A |