CN118206658A - Chimeric antigen receptor of targeted BCMA based on fully human and murine single chain antibodies and application thereof - Google Patents
Chimeric antigen receptor of targeted BCMA based on fully human and murine single chain antibodies and application thereof Download PDFInfo
- Publication number
- CN118206658A CN118206658A CN202410294964.5A CN202410294964A CN118206658A CN 118206658 A CN118206658 A CN 118206658A CN 202410294964 A CN202410294964 A CN 202410294964A CN 118206658 A CN118206658 A CN 118206658A
- Authority
- CN
- China
- Prior art keywords
- seq
- heavy chain
- light chain
- cells
- cdr2
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 title claims abstract description 89
- 108010008014 B-Cell Maturation Antigen Proteins 0.000 title claims abstract description 68
- 102000006942 B-Cell Maturation Antigen Human genes 0.000 title claims abstract description 67
- 241001529936 Murinae Species 0.000 title abstract description 17
- BGFTWECWAICPDG-UHFFFAOYSA-N 2-[bis(4-chlorophenyl)methyl]-4-n-[3-[bis(4-chlorophenyl)methyl]-4-(dimethylamino)phenyl]-1-n,1-n-dimethylbenzene-1,4-diamine Chemical compound C1=C(C(C=2C=CC(Cl)=CC=2)C=2C=CC(Cl)=CC=2)C(N(C)C)=CC=C1NC(C=1)=CC=C(N(C)C)C=1C(C=1C=CC(Cl)=CC=1)C1=CC=C(Cl)C=C1 BGFTWECWAICPDG-UHFFFAOYSA-N 0.000 title abstract 5
- 210000004027 cell Anatomy 0.000 claims abstract description 183
- 230000027455 binding Effects 0.000 claims abstract description 70
- 230000003834 intracellular effect Effects 0.000 claims abstract description 23
- 239000000427 antigen Substances 0.000 claims description 66
- 108091007433 antigens Proteins 0.000 claims description 66
- 102000036639 antigens Human genes 0.000 claims description 66
- 239000012634 fragment Substances 0.000 claims description 47
- 239000012642 immune effector Substances 0.000 claims description 40
- 229940121354 immunomodulator Drugs 0.000 claims description 40
- 206010028980 Neoplasm Diseases 0.000 claims description 39
- 201000011510 cancer Diseases 0.000 claims description 25
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 22
- 102000040430 polynucleotide Human genes 0.000 claims description 20
- 108091033319 polynucleotide Proteins 0.000 claims description 20
- 239000002157 polynucleotide Substances 0.000 claims description 20
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 20
- 229920001184 polypeptide Polymers 0.000 claims description 19
- 101000801255 Homo sapiens Tumor necrosis factor receptor superfamily member 17 Proteins 0.000 claims description 18
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 18
- 102000046935 human TNFRSF17 Human genes 0.000 claims description 15
- 238000011282 treatment Methods 0.000 claims description 13
- 238000004519 manufacturing process Methods 0.000 claims description 10
- 239000003814 drug Substances 0.000 claims description 6
- 230000036737 immune function Effects 0.000 claims description 5
- 230000004936 stimulating effect Effects 0.000 claims description 4
- 210000003719 b-lymphocyte Anatomy 0.000 abstract description 8
- 230000002147 killing effect Effects 0.000 abstract description 8
- 230000003211 malignant effect Effects 0.000 abstract description 4
- 230000008685 targeting Effects 0.000 abstract description 4
- 210000001744 T-lymphocyte Anatomy 0.000 description 72
- 239000013598 vector Substances 0.000 description 54
- 108090000623 proteins and genes Proteins 0.000 description 47
- 108091006020 Fc-tagged proteins Proteins 0.000 description 32
- 238000000034 method Methods 0.000 description 29
- 206010035226 Plasma cell myeloma Diseases 0.000 description 28
- 208000034578 Multiple myelomas Diseases 0.000 description 24
- 230000014509 gene expression Effects 0.000 description 21
- 108010076504 Protein Sorting Signals Proteins 0.000 description 20
- 239000008194 pharmaceutical composition Substances 0.000 description 19
- 230000011664 signaling Effects 0.000 description 18
- 241000700605 Viruses Species 0.000 description 17
- 238000002965 ELISA Methods 0.000 description 16
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 description 16
- 239000000243 solution Substances 0.000 description 16
- 238000006243 chemical reaction Methods 0.000 description 15
- 230000000139 costimulatory effect Effects 0.000 description 15
- 239000006228 supernatant Substances 0.000 description 15
- 108020004414 DNA Proteins 0.000 description 13
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 12
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 description 12
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 12
- 238000010276 construction Methods 0.000 description 12
- 239000012636 effector Substances 0.000 description 12
- 239000011550 stock solution Substances 0.000 description 12
- 230000006870 function Effects 0.000 description 11
- 239000013612 plasmid Substances 0.000 description 11
- 239000000047 product Substances 0.000 description 11
- 238000012216 screening Methods 0.000 description 11
- 101001023379 Homo sapiens Lysosome-associated membrane glycoprotein 1 Proteins 0.000 description 10
- 102100035133 Lysosome-associated membrane glycoprotein 1 Human genes 0.000 description 10
- 239000005018 casein Substances 0.000 description 10
- BECPQYXYKAMYBN-UHFFFAOYSA-N casein, tech. Chemical compound NCCCCC(C(O)=O)N=C(O)C(CC(O)=O)N=C(O)C(CCC(O)=N)N=C(O)C(CC(C)C)N=C(O)C(CCC(O)=O)N=C(O)C(CC(O)=O)N=C(O)C(CCC(O)=O)N=C(O)C(C(C)O)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=O)N=C(O)C(CCC(O)=O)N=C(O)C(COP(O)(O)=O)N=C(O)C(CCC(O)=N)N=C(O)C(N)CC1=CC=CC=C1 BECPQYXYKAMYBN-UHFFFAOYSA-N 0.000 description 10
- 235000021240 caseins Nutrition 0.000 description 10
- 239000013604 expression vector Substances 0.000 description 10
- 208000015181 infectious disease Diseases 0.000 description 10
- 230000004083 survival effect Effects 0.000 description 10
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 9
- 230000004913 activation Effects 0.000 description 9
- 239000011324 bead Substances 0.000 description 9
- 239000002299 complementary DNA Substances 0.000 description 9
- 238000002474 experimental method Methods 0.000 description 9
- 239000005090 green fluorescent protein Substances 0.000 description 9
- 150000007523 nucleic acids Chemical group 0.000 description 9
- 102000004127 Cytokines Human genes 0.000 description 8
- 108090000695 Cytokines Proteins 0.000 description 8
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 8
- 241000713666 Lentivirus Species 0.000 description 8
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 8
- 229960000723 ampicillin Drugs 0.000 description 8
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 8
- 238000003776 cleavage reaction Methods 0.000 description 8
- 238000001514 detection method Methods 0.000 description 8
- UQLDLKMNUJERMK-UHFFFAOYSA-L di(octadecanoyloxy)lead Chemical compound [Pb+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O UQLDLKMNUJERMK-UHFFFAOYSA-L 0.000 description 8
- 230000003053 immunization Effects 0.000 description 8
- 238000002649 immunization Methods 0.000 description 8
- 238000011534 incubation Methods 0.000 description 8
- 239000000203 mixture Substances 0.000 description 8
- 102000004169 proteins and genes Human genes 0.000 description 8
- 230000007017 scission Effects 0.000 description 8
- 230000028327 secretion Effects 0.000 description 8
- 238000011144 upstream manufacturing Methods 0.000 description 8
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 7
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 7
- 235000001014 amino acid Nutrition 0.000 description 7
- 238000003556 assay Methods 0.000 description 7
- 239000003153 chemical reaction reagent Substances 0.000 description 7
- 239000000499 gel Substances 0.000 description 7
- 230000004068 intracellular signaling Effects 0.000 description 7
- 239000003446 ligand Substances 0.000 description 7
- 239000002609 medium Substances 0.000 description 7
- 239000013642 negative control Substances 0.000 description 7
- 102000039446 nucleic acids Human genes 0.000 description 7
- 108020004707 nucleic acids Proteins 0.000 description 7
- 230000035755 proliferation Effects 0.000 description 7
- 238000011084 recovery Methods 0.000 description 7
- 210000002966 serum Anatomy 0.000 description 7
- 230000000638 stimulation Effects 0.000 description 7
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 6
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 6
- 108091007741 Chimeric antigen receptor T cells Proteins 0.000 description 6
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 6
- 102100037850 Interferon gamma Human genes 0.000 description 6
- 108010074328 Interferon-gamma Proteins 0.000 description 6
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 6
- 230000015572 biosynthetic process Effects 0.000 description 6
- 210000004369 blood Anatomy 0.000 description 6
- 239000008280 blood Substances 0.000 description 6
- 238000004113 cell culture Methods 0.000 description 6
- 230000010261 cell growth Effects 0.000 description 6
- 239000003795 chemical substances by application Substances 0.000 description 6
- 230000000694 effects Effects 0.000 description 6
- 210000002865 immune cell Anatomy 0.000 description 6
- 238000004806 packaging method and process Methods 0.000 description 6
- 239000002245 particle Substances 0.000 description 6
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 6
- 210000004180 plasmocyte Anatomy 0.000 description 6
- 238000002360 preparation method Methods 0.000 description 6
- 230000009870 specific binding Effects 0.000 description 6
- 239000000725 suspension Substances 0.000 description 6
- 238000003786 synthesis reaction Methods 0.000 description 6
- 239000013603 viral vector Substances 0.000 description 6
- 108010002350 Interleukin-2 Proteins 0.000 description 5
- 102000000588 Interleukin-2 Human genes 0.000 description 5
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- 102100029215 Signaling lymphocytic activation molecule Human genes 0.000 description 5
- 108010090804 Streptavidin Proteins 0.000 description 5
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 5
- 150000001413 amino acids Chemical class 0.000 description 5
- 238000009169 immunotherapy Methods 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- 238000001556 precipitation Methods 0.000 description 5
- 235000018102 proteins Nutrition 0.000 description 5
- 210000000952 spleen Anatomy 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 210000004881 tumor cell Anatomy 0.000 description 5
- 241000894006 Bacteria Species 0.000 description 4
- 102100024263 CD160 antigen Human genes 0.000 description 4
- 241000283707 Capra Species 0.000 description 4
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 4
- 101000761938 Homo sapiens CD160 antigen Proteins 0.000 description 4
- 101000633786 Homo sapiens SLAM family member 6 Proteins 0.000 description 4
- 101000633782 Homo sapiens SLAM family member 8 Proteins 0.000 description 4
- 102100032818 Integrin alpha-4 Human genes 0.000 description 4
- 102000003960 Ligases Human genes 0.000 description 4
- 108090000364 Ligases Proteins 0.000 description 4
- 102100029197 SLAM family member 6 Human genes 0.000 description 4
- 102100029214 SLAM family member 8 Human genes 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 4
- 102100040247 Tumor necrosis factor Human genes 0.000 description 4
- 102100028785 Tumor necrosis factor receptor superfamily member 14 Human genes 0.000 description 4
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 4
- 230000000259 anti-tumor effect Effects 0.000 description 4
- 239000012472 biological sample Substances 0.000 description 4
- 238000010367 cloning Methods 0.000 description 4
- 239000013599 cloning vector Substances 0.000 description 4
- 239000011248 coating agent Substances 0.000 description 4
- 238000000576 coating method Methods 0.000 description 4
- 238000010790 dilution Methods 0.000 description 4
- 239000012895 dilution Substances 0.000 description 4
- 238000010494 dissociation reaction Methods 0.000 description 4
- 230000005593 dissociations Effects 0.000 description 4
- 239000003937 drug carrier Substances 0.000 description 4
- 210000004962 mammalian cell Anatomy 0.000 description 4
- 230000007246 mechanism Effects 0.000 description 4
- 238000002156 mixing Methods 0.000 description 4
- 201000000050 myeloid neoplasm Diseases 0.000 description 4
- 238000000746 purification Methods 0.000 description 4
- 102000005962 receptors Human genes 0.000 description 4
- 108020003175 receptors Proteins 0.000 description 4
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 4
- 102000016605 B-Cell Activating Factor Human genes 0.000 description 3
- 108010028006 B-Cell Activating Factor Proteins 0.000 description 3
- 238000011357 CAR T-cell therapy Methods 0.000 description 3
- 101150013553 CD40 gene Proteins 0.000 description 3
- 102100032937 CD40 ligand Human genes 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 3
- 102100034459 Hepatitis A virus cellular receptor 1 Human genes 0.000 description 3
- 101001078158 Homo sapiens Integrin alpha-1 Proteins 0.000 description 3
- 101000994375 Homo sapiens Integrin alpha-4 Proteins 0.000 description 3
- 101000935040 Homo sapiens Integrin beta-2 Proteins 0.000 description 3
- 101000633778 Homo sapiens SLAM family member 5 Proteins 0.000 description 3
- 101000633784 Homo sapiens SLAM family member 7 Proteins 0.000 description 3
- 101000934346 Homo sapiens T-cell surface antigen CD2 Proteins 0.000 description 3
- 101000801234 Homo sapiens Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 description 3
- 101000679851 Homo sapiens Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 3
- 102100025323 Integrin alpha-1 Human genes 0.000 description 3
- 102100032816 Integrin alpha-6 Human genes 0.000 description 3
- 102100025390 Integrin beta-2 Human genes 0.000 description 3
- 108060001084 Luciferase Proteins 0.000 description 3
- 239000005089 Luciferase Substances 0.000 description 3
- 102100038082 Natural killer cell receptor 2B4 Human genes 0.000 description 3
- 102100029216 SLAM family member 5 Human genes 0.000 description 3
- 102100029198 SLAM family member 7 Human genes 0.000 description 3
- 108010074687 Signaling Lymphocytic Activation Molecule Family Member 1 Proteins 0.000 description 3
- 102100025237 T-cell surface antigen CD2 Human genes 0.000 description 3
- 102100033447 T-lymphocyte surface antigen Ly-9 Human genes 0.000 description 3
- 239000004098 Tetracycline Substances 0.000 description 3
- 102100036922 Tumor necrosis factor ligand superfamily member 13B Human genes 0.000 description 3
- 101710181056 Tumor necrosis factor ligand superfamily member 13B Proteins 0.000 description 3
- 102100033726 Tumor necrosis factor receptor superfamily member 17 Human genes 0.000 description 3
- 102100033728 Tumor necrosis factor receptor superfamily member 18 Human genes 0.000 description 3
- 230000003321 amplification Effects 0.000 description 3
- 238000000137 annealing Methods 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 230000022534 cell killing Effects 0.000 description 3
- 238000002659 cell therapy Methods 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 238000012258 culturing Methods 0.000 description 3
- 230000001086 cytosolic effect Effects 0.000 description 3
- 238000004925 denaturation Methods 0.000 description 3
- 230000036425 denaturation Effects 0.000 description 3
- 238000013461 design Methods 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 210000003527 eukaryotic cell Anatomy 0.000 description 3
- 238000012239 gene modification Methods 0.000 description 3
- 230000005017 genetic modification Effects 0.000 description 3
- 235000013617 genetically modified food Nutrition 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 230000002163 immunogen Effects 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 230000002458 infectious effect Effects 0.000 description 3
- 238000001802 infusion Methods 0.000 description 3
- 238000010369 molecular cloning Methods 0.000 description 3
- 230000004001 molecular interaction Effects 0.000 description 3
- 238000003199 nucleic acid amplification method Methods 0.000 description 3
- 210000004976 peripheral blood cell Anatomy 0.000 description 3
- 238000002823 phage display Methods 0.000 description 3
- 238000012257 pre-denaturation Methods 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 210000004989 spleen cell Anatomy 0.000 description 3
- 229960002180 tetracycline Drugs 0.000 description 3
- 229930101283 tetracycline Natural products 0.000 description 3
- 235000019364 tetracycline Nutrition 0.000 description 3
- 150000003522 tetracyclines Chemical class 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 230000009258 tissue cross reactivity Effects 0.000 description 3
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 2
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 2
- -1 AIM-V Substances 0.000 description 2
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 2
- 102100038080 B-cell receptor CD22 Human genes 0.000 description 2
- 102100040840 C-type lectin domain family 7 member A Human genes 0.000 description 2
- 102100038077 CD226 antigen Human genes 0.000 description 2
- 102100036008 CD48 antigen Human genes 0.000 description 2
- 102100027217 CD82 antigen Human genes 0.000 description 2
- 230000004544 DNA amplification Effects 0.000 description 2
- 102100025012 Dipeptidyl peptidase 4 Human genes 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 2
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 2
- 208000002250 Hematologic Neoplasms Diseases 0.000 description 2
- 229920000209 Hexadimethrine bromide Polymers 0.000 description 2
- 101000864344 Homo sapiens B- and T-lymphocyte attenuator Proteins 0.000 description 2
- 101000884305 Homo sapiens B-cell receptor CD22 Proteins 0.000 description 2
- 101000884298 Homo sapiens CD226 antigen Proteins 0.000 description 2
- 101000868215 Homo sapiens CD40 ligand Proteins 0.000 description 2
- 101000994365 Homo sapiens Integrin alpha-6 Proteins 0.000 description 2
- 101001046687 Homo sapiens Integrin alpha-E Proteins 0.000 description 2
- 101000935043 Homo sapiens Integrin beta-1 Proteins 0.000 description 2
- 101000971538 Homo sapiens Killer cell lectin-like receptor subfamily F member 1 Proteins 0.000 description 2
- 101000873418 Homo sapiens P-selectin glycoprotein ligand 1 Proteins 0.000 description 2
- 101000633792 Homo sapiens SLAM family member 9 Proteins 0.000 description 2
- 101000633780 Homo sapiens Signaling lymphocytic activation molecule Proteins 0.000 description 2
- 101000596234 Homo sapiens T-cell surface protein tactile Proteins 0.000 description 2
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 2
- 101000845170 Homo sapiens Thymic stromal lymphopoietin Proteins 0.000 description 2
- 101000795167 Homo sapiens Tumor necrosis factor receptor superfamily member 13B Proteins 0.000 description 2
- 101000795169 Homo sapiens Tumor necrosis factor receptor superfamily member 13C Proteins 0.000 description 2
- 101000648507 Homo sapiens Tumor necrosis factor receptor superfamily member 14 Proteins 0.000 description 2
- 101000679903 Homo sapiens Tumor necrosis factor receptor superfamily member 25 Proteins 0.000 description 2
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 2
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 2
- 108060003951 Immunoglobulin Proteins 0.000 description 2
- 102100021317 Inducible T-cell costimulator Human genes 0.000 description 2
- 102100022341 Integrin alpha-E Human genes 0.000 description 2
- 102100022297 Integrin alpha-X Human genes 0.000 description 2
- 102100025304 Integrin beta-1 Human genes 0.000 description 2
- 108010065805 Interleukin-12 Proteins 0.000 description 2
- 108020004684 Internal Ribosome Entry Sites Proteins 0.000 description 2
- 102100021458 Killer cell lectin-like receptor subfamily F member 1 Human genes 0.000 description 2
- 102000004083 Lymphotoxin-alpha Human genes 0.000 description 2
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 101100407308 Mus musculus Pdcd1lg2 gene Proteins 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- 101710141230 Natural killer cell receptor 2B4 Proteins 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 102100034925 P-selectin glycoprotein ligand 1 Human genes 0.000 description 2
- 108700030875 Programmed Cell Death 1 Ligand 2 Proteins 0.000 description 2
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 2
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 2
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 2
- 238000010802 RNA extraction kit Methods 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- 102100029196 SLAM family member 9 Human genes 0.000 description 2
- 230000006044 T cell activation Effects 0.000 description 2
- 108091008874 T cell receptors Proteins 0.000 description 2
- 102100035268 T-cell surface protein tactile Human genes 0.000 description 2
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 2
- 102100031294 Thymic stromal lymphopoietin Human genes 0.000 description 2
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 2
- 102100032101 Tumor necrosis factor ligand superfamily member 9 Human genes 0.000 description 2
- 102100029675 Tumor necrosis factor receptor superfamily member 13B Human genes 0.000 description 2
- 102100029690 Tumor necrosis factor receptor superfamily member 13C Human genes 0.000 description 2
- 102100022203 Tumor necrosis factor receptor superfamily member 25 Human genes 0.000 description 2
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 2
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 2
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 239000000654 additive Substances 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 2
- 230000006287 biotinylation Effects 0.000 description 2
- 238000007413 biotinylation Methods 0.000 description 2
- 210000004899 c-terminal region Anatomy 0.000 description 2
- 101150058049 car gene Proteins 0.000 description 2
- 238000002512 chemotherapy Methods 0.000 description 2
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 2
- 238000007796 conventional method Methods 0.000 description 2
- 239000012228 culture supernatant Substances 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 229940127089 cytotoxic agent Drugs 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 230000003013 cytotoxicity Effects 0.000 description 2
- 231100000135 cytotoxicity Toxicity 0.000 description 2
- 239000000539 dimer Substances 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 210000001671 embryonic stem cell Anatomy 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 230000005714 functional activity Effects 0.000 description 2
- 108020001507 fusion proteins Proteins 0.000 description 2
- 102000037865 fusion proteins Human genes 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 235000013922 glutamic acid Nutrition 0.000 description 2
- 239000004220 glutamic acid Substances 0.000 description 2
- 239000001963 growth medium Substances 0.000 description 2
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 2
- 102000018358 immunoglobulin Human genes 0.000 description 2
- 238000002513 implantation Methods 0.000 description 2
- 230000001965 increasing effect Effects 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 230000010354 integration Effects 0.000 description 2
- 229930027917 kanamycin Natural products 0.000 description 2
- 229960000318 kanamycin Drugs 0.000 description 2
- SBUJHOSQTJFQJX-NOAMYHISSA-N kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 description 2
- 229930182823 kanamycin A Natural products 0.000 description 2
- 238000012917 library technology Methods 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 230000007774 longterm Effects 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 210000003519 mature b lymphocyte Anatomy 0.000 description 2
- 239000000693 micelle Substances 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 210000000822 natural killer cell Anatomy 0.000 description 2
- 239000002773 nucleotide Substances 0.000 description 2
- 125000003729 nucleotide group Chemical group 0.000 description 2
- 238000004091 panning Methods 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 239000013600 plasmid vector Substances 0.000 description 2
- 210000001778 pluripotent stem cell Anatomy 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 210000001236 prokaryotic cell Anatomy 0.000 description 2
- 230000002062 proliferating effect Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 239000000523 sample Substances 0.000 description 2
- 230000003248 secreting effect Effects 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- JJGWLCLUQNFDIS-GTSONSFRSA-M sodium;1-[6-[5-[(3as,4s,6ar)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoylamino]hexanoyloxy]-2,5-dioxopyrrolidine-3-sulfonate Chemical compound [Na+].O=C1C(S(=O)(=O)[O-])CC(=O)N1OC(=O)CCCCCNC(=O)CCCC[C@H]1[C@H]2NC(=O)N[C@H]2CS1 JJGWLCLUQNFDIS-GTSONSFRSA-M 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 239000012089 stop solution Substances 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
- 239000000126 substance Substances 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 239000013589 supplement Substances 0.000 description 2
- 230000001502 supplementing effect Effects 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- JPSHPWJJSVEEAX-OWPBQMJCSA-N (2s)-2-amino-4-fluoranylpentanedioic acid Chemical compound OC(=O)[C@@H](N)CC([18F])C(O)=O JPSHPWJJSVEEAX-OWPBQMJCSA-N 0.000 description 1
- 108010082808 4-1BB Ligand Proteins 0.000 description 1
- QFVHZQCOUORWEI-UHFFFAOYSA-N 4-[(4-anilino-5-sulfonaphthalen-1-yl)diazenyl]-5-hydroxynaphthalene-2,7-disulfonic acid Chemical compound C=12C(O)=CC(S(O)(=O)=O)=CC2=CC(S(O)(=O)=O)=CC=1N=NC(C1=CC=CC(=C11)S(O)(=O)=O)=CC=C1NC1=CC=CC=C1 QFVHZQCOUORWEI-UHFFFAOYSA-N 0.000 description 1
- 206010003445 Ascites Diseases 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 206010006002 Bone pain Diseases 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 108010056102 CD100 antigen Proteins 0.000 description 1
- 108010017009 CD11b Antigen Proteins 0.000 description 1
- 102000007499 CD27 Ligand Human genes 0.000 description 1
- 108010046080 CD27 Ligand Proteins 0.000 description 1
- 102100027207 CD27 antigen Human genes 0.000 description 1
- 102100038078 CD276 antigen Human genes 0.000 description 1
- 102000004634 CD30 Ligand Human genes 0.000 description 1
- 108010017987 CD30 Ligand Proteins 0.000 description 1
- 108010029697 CD40 Ligand Proteins 0.000 description 1
- 108010038940 CD48 Antigen Proteins 0.000 description 1
- 108010084313 CD58 Antigens Proteins 0.000 description 1
- 108010062802 CD66 antigens Proteins 0.000 description 1
- 101710139831 CD82 antigen Proteins 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 101100288387 Caenorhabditis elegans lab-1 gene Proteins 0.000 description 1
- 102100024533 Carcinoembryonic antigen-related cell adhesion molecule 1 Human genes 0.000 description 1
- 102100025466 Carcinoembryonic antigen-related cell adhesion molecule 3 Human genes 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 1
- 102100027816 Cytotoxic and regulatory T-cell molecule Human genes 0.000 description 1
- 102000007260 Deoxyribonuclease I Human genes 0.000 description 1
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 206010061819 Disease recurrence Diseases 0.000 description 1
- 206010059866 Drug resistance Diseases 0.000 description 1
- 102100031984 Ephrin type-B receptor 6 Human genes 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 208000009329 Graft vs Host Disease Diseases 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 102100035943 HERV-H LTR-associating protein 2 Human genes 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 108010007712 Hepatitis A Virus Cellular Receptor 1 Proteins 0.000 description 1
- 101710185991 Hepatitis A virus cellular receptor 1 homolog Proteins 0.000 description 1
- 102100026122 High affinity immunoglobulin gamma Fc receptor I Human genes 0.000 description 1
- 101000971171 Homo sapiens Apoptosis regulator Bcl-2 Proteins 0.000 description 1
- 101000749325 Homo sapiens C-type lectin domain family 7 member A Proteins 0.000 description 1
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 1
- 101000716130 Homo sapiens CD48 antigen Proteins 0.000 description 1
- 101000914469 Homo sapiens CD82 antigen Proteins 0.000 description 1
- 101000914337 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 3 Proteins 0.000 description 1
- 101000908391 Homo sapiens Dipeptidyl peptidase 4 Proteins 0.000 description 1
- 101001064451 Homo sapiens Ephrin type-B receptor 6 Proteins 0.000 description 1
- 101001068136 Homo sapiens Hepatitis A virus cellular receptor 1 Proteins 0.000 description 1
- 101000913074 Homo sapiens High affinity immunoglobulin gamma Fc receptor I Proteins 0.000 description 1
- 101001056180 Homo sapiens Induced myeloid leukemia cell differentiation protein Mcl-1 Proteins 0.000 description 1
- 101001035237 Homo sapiens Integrin alpha-D Proteins 0.000 description 1
- 101001046683 Homo sapiens Integrin alpha-L Proteins 0.000 description 1
- 101001046668 Homo sapiens Integrin alpha-X Proteins 0.000 description 1
- 101001015037 Homo sapiens Integrin beta-7 Proteins 0.000 description 1
- 101000777628 Homo sapiens Leukocyte antigen CD37 Proteins 0.000 description 1
- 101000980823 Homo sapiens Leukocyte surface antigen CD53 Proteins 0.000 description 1
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 1
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 1
- 101001063392 Homo sapiens Lymphocyte function-associated antigen 3 Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101001109503 Homo sapiens NKG2-C type II integral membrane protein Proteins 0.000 description 1
- 101001109501 Homo sapiens NKG2-D type II integral membrane protein Proteins 0.000 description 1
- 101000589305 Homo sapiens Natural cytotoxicity triggering receptor 2 Proteins 0.000 description 1
- 101001098352 Homo sapiens OX-2 membrane glycoprotein Proteins 0.000 description 1
- 101000684208 Homo sapiens Prolyl endopeptidase FAP Proteins 0.000 description 1
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 1
- 101000914496 Homo sapiens T-cell antigen CD7 Proteins 0.000 description 1
- 101000837401 Homo sapiens T-cell leukemia/lymphoma protein 1A Proteins 0.000 description 1
- 101000837398 Homo sapiens T-cell leukemia/lymphoma protein 1B Proteins 0.000 description 1
- 101000946860 Homo sapiens T-cell surface glycoprotein CD3 epsilon chain Proteins 0.000 description 1
- 101000809875 Homo sapiens TYRO protein tyrosine kinase-binding protein Proteins 0.000 description 1
- 101000800116 Homo sapiens Thy-1 membrane glycoprotein Proteins 0.000 description 1
- 101000830596 Homo sapiens Tumor necrosis factor ligand superfamily member 15 Proteins 0.000 description 1
- 101000638251 Homo sapiens Tumor necrosis factor ligand superfamily member 9 Proteins 0.000 description 1
- 101000762805 Homo sapiens Tumor necrosis factor receptor superfamily member 19L Proteins 0.000 description 1
- 101000679857 Homo sapiens Tumor necrosis factor receptor superfamily member 3 Proteins 0.000 description 1
- 101000611185 Homo sapiens Tumor necrosis factor receptor superfamily member 5 Proteins 0.000 description 1
- 102100034980 ICOS ligand Human genes 0.000 description 1
- 102000017182 Ikaros Transcription Factor Human genes 0.000 description 1
- 108010013958 Ikaros Transcription Factor Proteins 0.000 description 1
- 102100026539 Induced myeloid leukemia cell differentiation protein Mcl-1 Human genes 0.000 description 1
- 102100023915 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 102100039904 Integrin alpha-D Human genes 0.000 description 1
- 102100022339 Integrin alpha-L Human genes 0.000 description 1
- 102100022338 Integrin alpha-M Human genes 0.000 description 1
- 108010041012 Integrin alpha4 Proteins 0.000 description 1
- 108010008212 Integrin alpha4beta1 Proteins 0.000 description 1
- 108010041100 Integrin alpha6 Proteins 0.000 description 1
- 108010030465 Integrin alpha6beta1 Proteins 0.000 description 1
- 102100033016 Integrin beta-7 Human genes 0.000 description 1
- 108090000174 Interleukin-10 Proteins 0.000 description 1
- 108090000172 Interleukin-15 Proteins 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 108010002586 Interleukin-7 Proteins 0.000 description 1
- PWKSKIMOESPYIA-BYPYZUCNSA-N L-N-acetyl-Cysteine Chemical compound CC(=O)N[C@@H](CS)C(O)=O PWKSKIMOESPYIA-BYPYZUCNSA-N 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
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-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
- 102000017578 LAG3 Human genes 0.000 description 1
- 101150030213 Lag3 gene Proteins 0.000 description 1
- 102100031586 Leukocyte antigen CD37 Human genes 0.000 description 1
- 102100024221 Leukocyte surface antigen CD53 Human genes 0.000 description 1
- 239000000232 Lipid Bilayer Substances 0.000 description 1
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 1
- 102100030984 Lymphocyte function-associated antigen 3 Human genes 0.000 description 1
- 108090000542 Lymphotoxin-alpha Proteins 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
- 101710085938 Matrix protein Proteins 0.000 description 1
- 101710127721 Membrane protein Proteins 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 101100493631 Mus musculus Bcl2l2 gene Proteins 0.000 description 1
- 101100328148 Mus musculus Cd300a gene Proteins 0.000 description 1
- 101000597780 Mus musculus Tumor necrosis factor ligand superfamily member 18 Proteins 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- 102100022683 NKG2-C type II integral membrane protein Human genes 0.000 description 1
- 102100022680 NKG2-D type II integral membrane protein Human genes 0.000 description 1
- 108010004217 Natural Cytotoxicity Triggering Receptor 1 Proteins 0.000 description 1
- 108010004222 Natural Cytotoxicity Triggering Receptor 3 Proteins 0.000 description 1
- 102100032870 Natural cytotoxicity triggering receptor 1 Human genes 0.000 description 1
- 102100032851 Natural cytotoxicity triggering receptor 2 Human genes 0.000 description 1
- 102100032852 Natural cytotoxicity triggering receptor 3 Human genes 0.000 description 1
- 102100037589 OX-2 membrane glycoprotein Human genes 0.000 description 1
- 102000004473 OX40 Ligand Human genes 0.000 description 1
- 108010042215 OX40 Ligand Proteins 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 101100377577 Picea glauca 3CAR gene Proteins 0.000 description 1
- 208000021161 Plasma cell disease Diseases 0.000 description 1
- 208000002151 Pleural effusion Diseases 0.000 description 1
- 102100039388 Polyamine deacetylase HDAC10 Human genes 0.000 description 1
- 101710107444 Polyamine deacetylase HDAC10 Proteins 0.000 description 1
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 1
- 102000003923 Protein Kinase C Human genes 0.000 description 1
- 108090000315 Protein Kinase C Proteins 0.000 description 1
- 238000001190 Q-PCR Methods 0.000 description 1
- 102000018795 RELT Human genes 0.000 description 1
- 108010052562 RELT Proteins 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 1
- 208000001647 Renal Insufficiency Diseases 0.000 description 1
- 102100027744 Semaphorin-4D Human genes 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 1
- 108700015968 Slam family Proteins 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 102100027208 T-cell antigen CD7 Human genes 0.000 description 1
- 102100039367 T-cell immunoglobulin and mucin domain-containing protein 4 Human genes 0.000 description 1
- 101710174757 T-cell immunoglobulin and mucin domain-containing protein 4 Proteins 0.000 description 1
- 102100028676 T-cell leukemia/lymphoma protein 1A Human genes 0.000 description 1
- 102100028678 T-cell leukemia/lymphoma protein 1B Human genes 0.000 description 1
- 102100035794 T-cell surface glycoprotein CD3 epsilon chain Human genes 0.000 description 1
- 101710114141 T-lymphocyte surface antigen Ly-9 Proteins 0.000 description 1
- 101150052863 THY1 gene Proteins 0.000 description 1
- 102100038717 TYRO protein tyrosine kinase-binding protein Human genes 0.000 description 1
- 108020005038 Terminator Codon Proteins 0.000 description 1
- 102100033523 Thy-1 membrane glycoprotein Human genes 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- 108010065158 Tumor Necrosis Factor Ligand Superfamily Member 14 Proteins 0.000 description 1
- 102100024586 Tumor necrosis factor ligand superfamily member 14 Human genes 0.000 description 1
- 102100024587 Tumor necrosis factor ligand superfamily member 15 Human genes 0.000 description 1
- 102100026716 Tumor necrosis factor receptor superfamily member 19L Human genes 0.000 description 1
- 102100033733 Tumor necrosis factor receptor superfamily member 1B Human genes 0.000 description 1
- 101710187830 Tumor necrosis factor receptor superfamily member 1B Proteins 0.000 description 1
- 102100022156 Tumor necrosis factor receptor superfamily member 3 Human genes 0.000 description 1
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 1
- 206010046865 Vaccinia virus infection Diseases 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 229960004308 acetylcysteine Drugs 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 238000000246 agarose gel electrophoresis Methods 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- OFHCOWSQAMBJIW-AVJTYSNKSA-N alfacalcidol Chemical compound C1(/[C@@H]2CC[C@@H]([C@]2(CCC1)C)[C@H](C)CCCC(C)C)=C\C=C1\C[C@@H](O)C[C@H](O)C1=C OFHCOWSQAMBJIW-AVJTYSNKSA-N 0.000 description 1
- 230000000735 allogeneic effect Effects 0.000 description 1
- 229940024606 amino acid Drugs 0.000 description 1
- 208000007502 anemia Diseases 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000006909 anti-apoptosis Effects 0.000 description 1
- 230000006023 anti-tumor response Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 229940125644 antibody drug Drugs 0.000 description 1
- 210000000612 antigen-presenting cell Anatomy 0.000 description 1
- 239000002246 antineoplastic agent Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 230000005975 antitumor immune response Effects 0.000 description 1
- 238000002617 apheresis Methods 0.000 description 1
- 230000001640 apoptogenic effect Effects 0.000 description 1
- 230000005775 apoptotic pathway Effects 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 239000000823 artificial membrane Substances 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 108010010804 beta2 Heterotrimer Lymphotoxin alpha1 Proteins 0.000 description 1
- 238000010170 biological method Methods 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 229960005520 bryostatin Drugs 0.000 description 1
- MJQUEDHRCUIRLF-TVIXENOKSA-N bryostatin 1 Chemical compound C([C@@H]1CC(/[C@@H]([C@@](C(C)(C)/C=C/2)(O)O1)OC(=O)/C=C/C=C/CCC)=C\C(=O)OC)[C@H]([C@@H](C)O)OC(=O)C[C@H](O)C[C@@H](O1)C[C@H](OC(C)=O)C(C)(C)[C@]1(O)C[C@@H]1C\C(=C\C(=O)OC)C[C@H]\2O1 MJQUEDHRCUIRLF-TVIXENOKSA-N 0.000 description 1
- MUIWQCKLQMOUAT-AKUNNTHJSA-N bryostatin 20 Natural products COC(=O)C=C1C[C@@]2(C)C[C@]3(O)O[C@](C)(C[C@@H](O)CC(=O)O[C@](C)(C[C@@]4(C)O[C@](O)(CC5=CC(=O)O[C@]45C)C(C)(C)C=C[C@@](C)(C1)O2)[C@@H](C)O)C[C@H](OC(=O)C(C)(C)C)C3(C)C MUIWQCKLQMOUAT-AKUNNTHJSA-N 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000003710 calcium ionophore 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
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 229920006317 cationic polymer Polymers 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000006037 cell lysis Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 239000003638 chemical reducing agent Substances 0.000 description 1
- 108010072917 class-I restricted T cell-associated molecule Proteins 0.000 description 1
- 238000001246 colloidal dispersion Methods 0.000 description 1
- 239000000084 colloidal system Substances 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- NKLPQNGYXWVELD-UHFFFAOYSA-M coomassie brilliant blue Chemical compound [Na+].C1=CC(OCC)=CC=C1NC1=CC=C(C(=C2C=CC(C=C2)=[N+](CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=2C=CC(=CC=2)N(CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=C1 NKLPQNGYXWVELD-UHFFFAOYSA-M 0.000 description 1
- 239000008358 core component Substances 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 239000002254 cytotoxic agent Substances 0.000 description 1
- 231100000599 cytotoxic agent Toxicity 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000009849 deactivation Effects 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 108010025838 dectin 1 Proteins 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000002405 diagnostic procedure Methods 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 238000006911 enzymatic reaction Methods 0.000 description 1
- 210000003979 eosinophil Anatomy 0.000 description 1
- CJAONIOAQZUHPN-KKLWWLSJSA-N ethyl 12-[[2-[(2r,3r)-3-[2-[(12-ethoxy-12-oxododecyl)-methylamino]-2-oxoethoxy]butan-2-yl]oxyacetyl]-methylamino]dodecanoate Chemical compound CCOC(=O)CCCCCCCCCCCN(C)C(=O)CO[C@H](C)[C@@H](C)OCC(=O)N(C)CCCCCCCCCCCC(=O)OCC CJAONIOAQZUHPN-KKLWWLSJSA-N 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 210000004700 fetal blood Anatomy 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- IJJVMEJXYNJXOJ-UHFFFAOYSA-N fluquinconazole Chemical compound C=1C=C(Cl)C=C(Cl)C=1N1C(=O)C2=CC(F)=CC=C2N=C1N1C=NC=N1 IJJVMEJXYNJXOJ-UHFFFAOYSA-N 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 238000003209 gene knockout Methods 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 208000024908 graft versus host disease Diseases 0.000 description 1
- 210000002443 helper t lymphocyte Anatomy 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 210000004408 hybridoma Anatomy 0.000 description 1
- 206010020718 hyperplasia Diseases 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 238000012151 immunohistochemical method Methods 0.000 description 1
- 239000002955 immunomodulating agent Substances 0.000 description 1
- 229960003444 immunosuppressant agent Drugs 0.000 description 1
- 230000001861 immunosuppressant effect Effects 0.000 description 1
- 239000003018 immunosuppressive agent Substances 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 108010043603 integrin alpha4beta7 Proteins 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 230000002601 intratumoral effect Effects 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 201000006370 kidney failure Diseases 0.000 description 1
- 208000021601 lentivirus infection Diseases 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 230000005923 long-lasting effect Effects 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 208000019420 lymphoid neoplasm Diseases 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 239000004005 microsphere Substances 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 239000002088 nanocapsule Substances 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 230000002688 persistence Effects 0.000 description 1
- 238000009520 phase I clinical trial Methods 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 238000000053 physical method Methods 0.000 description 1
- 230000008288 physiological mechanism Effects 0.000 description 1
- 108010058237 plasma protein fraction Proteins 0.000 description 1
- 229940002993 plasmanate Drugs 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 208000012584 pre-descemet corneal dystrophy Diseases 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 230000002797 proteolythic effect Effects 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 230000002000 scavenging effect Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 108091006024 signal transducing proteins Proteins 0.000 description 1
- 102000034285 signal transducing proteins Human genes 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 102000035025 signaling receptors Human genes 0.000 description 1
- 108091005475 signaling receptors Proteins 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 229940054269 sodium pyruvate Drugs 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000011272 standard treatment Methods 0.000 description 1
- 238000011146 sterile filtration Methods 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 101150065190 term gene Proteins 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
- 210000001541 thymus gland Anatomy 0.000 description 1
- 230000008791 toxic response Effects 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 238000011269 treatment regimen Methods 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
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 208000007089 vaccinia Diseases 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 238000012795 verification Methods 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2878—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
-
- 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
-
- 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
- A61K39/0005—Vertebrate antigens
- A61K39/0011—Cancer antigens
- A61K39/001102—Receptors, cell surface antigens or cell surface determinants
-
- 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
- A61K39/463—Cellular immunotherapy characterised by recombinant expression
- A61K39/4631—Chimeric Antigen Receptors [CAR]
-
- 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
- A61K39/464402—Receptors, cell surface antigens or cell surface determinants
- A61K39/464416—Receptors for cytokines
- A61K39/464417—Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K19/00—Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
-
- 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
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/62—DNA sequences coding for fusion proteins
-
- 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
- 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
- C12N15/867—Retroviral 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
- C12N7/00—Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
-
- 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
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/515—Animal cells
- A61K2039/5156—Animal cells expressing foreign proteins
-
- 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
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/515—Animal cells
- A61K2039/5158—Antigen-pulsed cells, e.g. T-cells
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/565—Complementarity determining region [CDR]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/02—Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/03—Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/33—Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
-
- 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
- C12N2510/00—Genetically modified cells
-
- 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
- C12N2740/15011—Lentivirus, not HIV, e.g. FIV, SIV
- C12N2740/15041—Use of virus, viral particle or viral elements as a vector
- C12N2740/15043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Zoology (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Biochemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Biotechnology (AREA)
- Cell Biology (AREA)
- Epidemiology (AREA)
- Mycology (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Virology (AREA)
- Plant Pathology (AREA)
- Oncology (AREA)
- Physics & Mathematics (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Hematology (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Peptides Or Proteins (AREA)
Abstract
The present invention relates to single chain antibodies targeting BCMA and chimeric antigen receptors based thereon and their uses. The present invention provides fully human and murine single chain antibodies that specifically bind to BCMA. The chimeric antigen receptor comprises an anti-BCMA fully human or murine single chain antibody, a human CD8 alpha hinge region, a human CD8 alpha transmembrane region, a human 41BB intracellular region and a human CD3 zeta intracellular region which are connected in sequence. Further, a CAR-T cell based on an anti-BCMA fully human or murine single chain antibody is provided that is capable of specifically binding to BCMA and effectively killing B lymphocyte lineage malignant cells that express BCMA.
Description
The invention relates to a chimeric antigen receptor targeting BCMA based on fully human and murine single chain antibodies and application thereof, and a divisional application of the invention patent application with the application number of CN 2021109121618.
Technical Field
The invention belongs to the field of antibodies and chimeric antigen receptors, and in particular relates to an antibody and chimeric antigen receptor targeting an extracellular segment of BCMA and application thereof.
Background
Multiple myeloma is a malignant plasma cell disease, which is manifested by malignant clonal hyperplasia of bone marrow plasma cells, secretion of monoclonal immunoglobulin or fragments thereof (M protein), which causes injury to relevant target organs or tissues such as bones and kidneys, and is frequently manifested clinically by bone pain, anemia, renal insufficiency, infection, etc. [ N Engl J Med,2011.364 (11): 1046-60]. At present, multiple myeloma is the second largest malignant tumor of the blood system, accounts for 10% of malignant tumors of the blood system, frequently occurs in men, the incidence rate of the multiple myeloma increases year by year with the age, and the multiple myeloma has a tendency of younger in recent years [ CA CancerJ Clin,2014.64 (1): 9-29]. The B cell maturation antigen (B-cell maturation antigen, BCMA), also called CD269, consists of 184 amino acid residues, the intracellular region of which contains 80 amino acid residues, the extracellular region sequence is very short, and only one carbohydrate recognition domain is the B cell surface molecule. BCMA, a type I transmembrane signaling protein lacking a signal peptide, is a member of the tumor necrosis factor receptor family (TNFR) that binds to both B cell activating factor BAFF or proliferation-inducing ligand (aproliferation induced ligand, APRIL) [ Curr Opin Struct Biol,2004.14 (2): 154-60]. In normal tissues, BCMA is expressed on the surface of mature B cells and plasma cells, the immune system of a BCMA gene knockout mouse is normal, has normal spleen structure, and the development of B lymphocytes is normal, but the number of plasma cells is obviously reduced, so that BCMA plays an important role in maintaining the survival of the plasma cells, and the mechanism mainly comprises BCMA BAFF protein binding, up-regulates anti-apoptosis genes Bcl-2, mcl-1, bclw and the like, and maintains cell growth [ J Exp Med,2004.199 (1): 91-8]. Similarly, this mechanism also functions in myeloma cells, playing an important role in promoting malignant proliferation of myeloma cells [ Blood,2004.103 (8): 3148-57]. Studies have shown that BCMA is ubiquitously expressed in multiple myeloma cell lines and that detection in multiple myeloma patients also yields consistent results [ Blood,2004.103 (2): 689-94]. Kochenderfer, et al, have studied in depth the expression profile of BCMA on the basis of the reported Q-PCR, flow cytometry and immunohistochemical methods, confirming that BCMA is not expressed in normal human tissues other than mature B cells, plasma cells, and also in cd34+ hematopoietic cells [ CLIN CANCER RES,2013.19 (8): 2048-60]. The high similarity of binding BCMA expression profile to CD19, and the successful progression of anti-CD 19 CAR-T cell therapy, suggest that we BCMA can be used as one of the targets of CAR-T cells for cellular immunotherapy of multiple myeloma. Chimeric antigen Receptor-T cells (CHIMERIC ANTIGEN Receptor-T cells, CAR-T) refer to T cells that, after genetic modification, recognize a specific antigen of interest in an MHC non-limiting manner and continue to activate expansion. The annual meeting of the international cell therapy association in 2012 indicates that biological immune cell therapy has become a fourth means for treating tumors outside surgery, radiotherapy and chemotherapy, and is becoming an essential means for future tumor treatment. CAR-T cell feedback therapy is the most clearly effective form of immunotherapy in current tumor therapy. A large number of researches show that the CAR-T cells can effectively recognize tumor antigens, cause specific anti-tumor immune response and obviously improve the survival condition of patients. Chimeric Antigen Receptors (CARs) are the core component of CAR-T, conferring to T cells the ability to recognize tumor antigens in an HLA-independent manner, which enables CAR engineered T cells to recognize a broader range of targets than native T cell surface receptor TCRs. The basic design of a CAR includes a Tumor Associated Antigen (TAA) binding region (typically an scFv fragment derived from a monoclonal antibody antigen binding region), an extracellular hinge region, a transmembrane region and an intracellular signaling region. The choice of antigen of interest is a critical determinant of the specificity, effectiveness of the CAR and safety of the genetically engineered T cells themselves.
With the continuous development of CAR-T technology, CAR-T is currently divided into four generations. First generation CAR-T cells consist of extracellular binding region-single chain antibodies (SINGLE CHAIN FRAGMENT variable, scFv), transmembrane region (transmembrane region, TM) and intracellular signaling region-immune receptor tyrosine-activating motif (immunoreceptor tyrosine based activation motif, ITAM), wherein the chimeric antigen receptor portions are linked as follows: scFv-TM-CD3 zeta.
Although some specific cytotoxicity can be seen in the first generation of CARs, the clinical trial summary of the first generation of CARs in 2006 shows poor efficacy. The reason for this is that the first generation of CAR-T cells are rapidly depleted in patients and have so poor persistence that CAR-T cells have been apoptotic to a large number of tumor cells that they can elicit an anti-tumor cytotoxic effect, but the cytokine secretion is relatively low, but their survival in vivo is short and not eliciting a long lasting anti-tumor effect [ CANCER RES,2007.67 (22): 11029-11036].
Second generation CAR-T cells optimize CAR design, the T cell activation signaling region remains a hotspot for research. Complete activation of T cells depends on the actions of dual signaling and cytokines. Wherein the first signal is a specific signal initiated by the TCR recognizing an antigen peptide-MHC complex on the surface of an antigen presenting cell; the second signal is a co-stimulatory signal. The second generation CAR [ J Immunol,1998.161 (6) appeared as early as 1998: 2791-7]. The generation 2 CAR adds a co-stimulatory molecule in the intracellular signal peptide region, namely, the co-stimulatory signal is assembled into the CAR, so that an activation signal can be better provided for the CAR-T cell, and the CAR can activate the co-stimulatory molecule and the intracellular signal at the same time after recognizing tumor cells, so that double activation is realized, and the proliferation secretion capacity and the anti-tumor effect of the T cell can be obviously improved. The first T cell costimulatory signaling receptor studied in detail was CD28, which is capable of binding to a B7 family member on the surface of target cells. Co-stimulation of CD28 promotes proliferation of T cells, synthesis and expression of IL-2, and enhances the ability of T cells to resist apoptosis. Co-stimulatory molecules such as CD134 (OX 40) and 41BB (4-1 BB) are then presented to enhance T cell cytotoxicity, proliferative activity, maintain T cell responses, extend T cell survival time, etc. Such second generation CARs produced unexpected effects in subsequent clinical trials, frequently triggering shocks based on clinical reports of second generation CARs since 2010, especially for relapsed, refractory ALL patients, with complete remission rates of up to 90% or more.
The third generation CAR signal peptide region integrates more than 2 co-stimulatory molecules, so that T cells can be continuously activated and proliferated, cytokines can be continuously secreted, and the capability of killing tumor cells by the T cells is more remarkable, namely, a stronger anti-tumor response can be obtained by the novel generation CAR [ Mol Ther,2005.12 (5): 933-941]. Most typically, a 41BB stimulus is added under the influence of the CD28 stimulus.
The fourth generation of CAR-T cells, with the addition of cytokines or co-stimulatory ligands, e.g., the fourth generation of CARs, can produce IL-12, which can modulate the immune microenvironment-increase T cell activation while activating innate immune cells to act to clear target antigen-negative cancer cells, thereby achieving a bi-regulatory effect [ Expert Opin Biol Ther,2015.1 (8): 1145-54].
One great advantage of CAR-T cells is that they are active drugs, and once infused, physiological mechanisms regulate T cell balance, memory formation, and antigen-driven expansion. However, this treatment is not perfect and T cells can be off target and attack other tissues, or the amount of expansion is too high to be desirable for treatment. Given that CAR-T cells have been included in standard therapeutic ranges, it is very useful to design a patient or drug-controlled activation or deactivation mechanism to regulate the presence of CAR-T cells. For technical reasons, the closing mechanism is more easily applied to T cells. As one of these, the iCas9 system is under clinical investigation. When the cells express the iCas9, small molecular compounds can induce the iCas9 precursor molecules to form dimers, and activate apoptosis pathways, so that the purpose of eliminating the cells is achieved. In graft versus host disease, small molecule AP1903 has been used to induce iCas9 dimers and clear T cells, suggesting the feasibility of this approach [ CLIN CANCER RES,2016.22 (8): 1875-84].
In addition, CAR-T cells can also be made to express proteins to which these antibodies are directed simultaneously, such as tgfr, using already clinically used scavenging antibodies, by administering antibody drugs to scavenge the corresponding CAR-T cells after the treatment-related toxic response has occurred or after the treatment has been completed [ SCI TRANSL MED,2015.7:275ra22].
9 Months 2015, carl June's J.In New England published an article of successful treatment of 1 patient with relapsed refractory Multiple Myeloma (MM) using CD19 molecular targeted CAR-T cell therapy [ NEJM,2015.373 (11): 1040-7]. Although multiple myeloma as a B cell line tumor does not normally express CD19, CD19 is not a target for multiple myeloma immunotherapy. A trace of multiple myeloma clones with drug resistance and disease recurrence characteristics have been reported to have a B cell phenotype (i.e., CD19 positive). After defining BCMA as a target of CAR-T cells, the national cancer institute Kochenderfer and the like successfully constructed anti-BCMACAR-T cells, and preclinical studies show that the CAR-T cells specifically recognize BCMA, and greatly expand and secrete cytokines and exert killing functions after being activated by BCMA, and also have anti-tumor effects in a mouse tumor model [ CLIN CANCER RES,2013.19 (8): 2048-60]. A phase I clinical study was conducted by the national cancer institute in 2014 against BCMACAR-T cells to treat multiple myeloma, and the clinical safety and efficacy of the anti BCMACAR-T cells was verified against patients with multiple myeloma who were unresponsive to the current standard treatment regimen (clinical Trials gov Identifier: NCT 02215967). The clinical trial results of CAR-T cell therapy phase I for patients with multiple myeloma were reported by the american national cancer institute medical oncology Syed Abbas Ali professor team at the 57 th american annual meeting at 12 months of 2015. The study is incorporated into 12 patients with refractory relapsing multiple myeloma who fail more than 3 lines of chemotherapy, and some patients have more than or equal to 50% of myeloma cells in the bone marrow. After BCMACAR-T cells were infused into these patients, 1 patient was completely relieved, 3 patients were partially relieved, and the rest were stable, thus the anti-BCMACAR-T cell therapy was first demonstrated to be effective against multiple myeloma without significant side effects, and was evaluated as one of the most influential clinical studies in ASH years [ Late-BreakingAbstracts, meeting abstract No.: LAB-1]. Currently, the ibutglom university of pennsylvania ibutglom cancer center has also registered a phase I clinical trial against BCMACAR-T cells for the treatment of multiple myeloma and was under development in the dense drum (clinical trimals gov Identifier: NCT 02546167).
Summary of The Invention
A plurality of humanized antibodies are obtained as targeting molecules on the surface of chimeric antigen receptor T cells by utilizing a phage display technology-based humanized natural antibody library technology. Similar work was also performed using a murine immune antibody library technology based on phage display technology. An anti-BCMA chimeric antigen receptor constructed based on an anti-BCMA single chain antibody (e.g., scFv), including murine and human (particularly HKl a) is provided that includes an anti-BCMA binding region, a human CD 8a hinge region, a human CD 8a transmembrane region, a human 41BB intracellular region, and a human cd3ζ intracellular region. T cells expressing this chimeric antigen receptor (especially HK 10-based) have the ability to specifically kill BCMA positive tumor cells and are better in terms of ifnγ secretion and CD107a expression than T cells expressing chimeric antigen receptor based on the c11d5.3 clone. T cells expressing the chimeric antigen receptor have better clinical treatment effect. In addition to specific tumor cell killing capacity, superior ifnγ secretion and CD107a expression capacity, when T cells carrying the chimeric antigen receptor (particularly HK 10-based) are used in clinical experiments, immunogenicity can be minimized due to the use of fully human antibody scFv, avoiding clearance of T cells carrying the chimeric antigen receptor, potentially prolonging the survival and improving therapeutic efficacy of T cells carrying the chimeric antigen receptor.
In one aspect, the invention relates to an antibody or antigen binding fragment that specifically binds to human B Cell Maturation Antigen (BCMA), characterized by comprising a heavy chain variable domain and a light chain variable domain, said comprising a heavy chain variable domain and a light chain variable domain:
(1) SEQ ID NO:1, a heavy chain CDR1, SEQ ID NO:2, a heavy chain CDR2, SEQ ID NO:3, a heavy chain CDR3, SEQ ID NO:4, light chain CDR1, SEQ ID NO:5 and the light chain CDR2 shown in SEQ ID NO:6, a light chain CDR3;
(2) SEQ ID NO:7, heavy chain CDR1, SEQ ID NO:8, a heavy chain CDR2, SEQ ID NO:9, heavy chain CDR3, SEQ ID NO:10, light chain CDR1, SEQ ID NO:11 and the light chain CDR2 and SEQ ID NO:12, a light chain CDR3;
(3) SEQ ID NO:13, a heavy chain CDR1, SEQ ID NO:14, a heavy chain CDR2, SEQ ID NO:15, a heavy chain CDR3, SEQ ID NO:16, light chain CDR1, SEQ ID NO:17 and the light chain CDR2 and SEQ ID NO:18, a light chain CDR3;
(4) SEQ ID NO:19, a heavy chain CDR1, SEQ ID NO:20, a heavy chain CDR2, SEQ ID NO:21, heavy chain CDR3, SEQ ID NO:22, light chain CDR1, SEQ ID NO:23 and the light chain CDR2 shown in SEQ ID NO:24, a light chain CDR3; or (b)
(5) SEQ ID NO:25, heavy chain CDRl, SEQ ID NO:26, a heavy chain CDR2, SEQ ID NO:27, heavy chain CDR3, SEQ ID NO:28, light chain CDR1, SEQ ID NO:29 and the light chain CDR2 and SEQ ID NO:30, and a light chain CDR3.
Preferably, the antibody or antigen binding fragment comprises SEQ ID NO:1, a heavy chain CDR1, SEQ ID NO:2, a heavy chain CDR2, SEQ ID NO:3, a heavy chain CDR3, SEQ ID NO:4, light chain CDR1, SEQ ID NO:5 and the light chain CDR2 shown in SEQ ID NO:6, a light chain CDR3.
In one embodiment of the invention, the antibody or antigen binding fragment is chimeric or human.
In another aspect, the invention relates to an antibody or antigen binding fragment that specifically binds to human B Cell Maturation Antigen (BCMA), characterized by comprising a heavy chain variable domain and a light chain variable domain comprising:
(1) SEQ ID NO:31, a heavy chain CDR1, SEQ ID NO:32, a heavy chain CDR2, SEQ ID NO:33, a heavy chain CDR3, SEQ ID NO:34, light chain CDR1, SEQ ID NO:35 and the light chain CDR2 and SEQ ID NO:36, a light chain CDR3;
(2) SEQ ID NO:37, a heavy chain CDR1, SEQ ID NO:38, the heavy chain CDR2, SEQ ID NO:39, heavy chain CDR3, SEQ ID NO:40, light chain CDR1, SEQ ID NO:41 and the light chain CDR2 and SEQ ID NO:42, a light chain CDR3;
(3) SEQ ID NO:43, a heavy chain CDR1, SEQ ID NO:44, a heavy chain CDR2, SEQ ID NO:45, heavy chain CDR3, SEQ ID NO:46, light chain CDR1, SEQ ID NO:47 and the light chain CDR2 and SEQ ID NO:48, a light chain CDR3;
(4) SEQ ID NO:49, heavy chain CDR1, SEQ ID NO:50, a heavy chain CDR2, SEQ ID NO:51, heavy chain CDR3, SEQ ID NO:52, light chain CDR1, SEQ ID NO:53 and the light chain CDR2 and SEQ ID NO:54 light chain CDR3; or (b)
(5) SEQ ID NO:55, heavy chain CDR1, SEQ ID NO:56, heavy chain CDR2, SEQ ID NO:57, a heavy chain CDR3, SEQ ID NO:58, light chain CDR1, SEQ ID NO:59 and light chain CDR2 and SEQ ID NO:60, and a light chain CDR3.
In one embodiment of the invention, the antibody or antigen binding fragment is murine, chimeric or humanized.
In one embodiment of the invention, the antibody or antigen binding fragment specifically binds to the extracellular domain of human B Cell Maturation Antigen (BCMA).
In one embodiment of the invention, the antibody or antigen binding fragment is a scFv, fab or whole antibody. In one embodiment of the invention, the heavy chain of the antibody or antigen binding fragment is IgG, in particular IgG 1. In one embodiment of the invention, the light chain of the antibody or antigen binding fragment is kappa (kappa) or lambda (lambda), in particular kappa.
In a further aspect, the invention relates to one or more polynucleotides, characterized in that it encodes an antibody or antigen binding fragment according to the invention.
In a further aspect, the invention relates to a vector, characterized in that it comprises a polynucleotide according to the invention. In one embodiment of the invention, the vector is a cloning vector or an expression vector, in particular a plasmid vector.
In a further aspect, the invention relates to a host cell, characterized in that it comprises a polynucleotide according to the invention or a vector according to the invention. In one embodiment of the invention, the host cell is a prokaryotic cell, in particular an E.coli cell. In another embodiment of the invention, the host cell is a eukaryotic cell, in particular a chinese hamster ovary cell.
In a further aspect, the invention relates to a method of producing an antibody or antigen-binding fragment, characterized in that a host cell according to the invention is cultured under conditions for expression of the antibody or antigen-binding fragment.
In a further aspect, the invention relates to a pharmaceutical composition characterized by comprising an antibody or antigen-binding fragment according to the invention, and one or more pharmaceutically acceptable carriers.
In yet another aspect, the invention relates to a chimeric antigen receptor characterized by comprising an extracellular region, a transmembrane region, and an intracellular region, wherein the extracellular region comprises an antigen binding region comprising a heavy chain variable domain and a light chain variable domain comprising:
(1) SEQ ID NO:1, a heavy chain CDR1, SEQ ID NO:2, a heavy chain CDR2, SEQ ID NO:3, a heavy chain CDR3, SEQ ID NO:4, light chain CDR1, SEQ ID NO:5 and the light chain CDR2 shown in SEQ ID NO:6, a light chain CDR3;
(2) SEQ ID NO:7, heavy chain CDR1, SEQ ID NO:8, a heavy chain CDR2, SEQ ID NO:9, heavy chain CDR3, SEQ ID NO:10, light chain CDR1, SEQ ID NO:11 and the light chain CDR2 and SEQ ID NO:12, a light chain CDR3;
(3) SEQ ID NO:13, a heavy chain CDR1, SEQ ID NO:14, a heavy chain CDR2, SEQ ID NO:15, a heavy chain CDR3, SEQ ID NO:16, light chain CDR1, SEQ ID NO:17 and the light chain CDR2 and SEQ ID NO:18, a light chain CDR3;
(4) SEQ ID NO:19, a heavy chain CDR1, SEQ ID NO:20, a heavy chain CDR2, SEQ ID NO:21, heavy chain CDR3, SEQ ID NO:22, light chain CDR1, SEQ ID NO:23 and the light chain CDR2 shown in SEQ ID NO:24, a light chain CDR3;
(5) SEQ ID NO:25, heavy chain CDRl, SEQ ID NO:26, a heavy chain CDR2, SEQ ID NO:27, heavy chain CDR3, SEQ ID NO:28, light chain CDR1, SEQ ID NO:29 and the light chain CDR2 and SEQ ID NO:30, a light chain CDR3;
(6) SEQ ID NO:31, a heavy chain CDR1, SEQ ID NO:32, a heavy chain CDR2, SEQ ID NO:33, a heavy chain CDR3, SEQ ID NO:34, light chain CDR1, SEQ ID NO:35 and the light chain CDR2 and SEQ ID NO:36, a light chain CDR3;
(7) SEQ ID NO:37, a heavy chain CDR1, SEQ ID NO:38, the heavy chain CDR2, SEQ ID NO:39, heavy chain CDR3, SEQ ID NO:40, light chain CDR1, SEQ ID NO:41 and the light chain CDR2 and SEQ ID NO:42, a light chain CDR3;
(8) SEQ ID NO:43, a heavy chain CDR1, SEQ ID NO:44, a heavy chain CDR2, SEQ ID NO:45, heavy chain CDR3, SEQ ID NO:46, light chain CDR1, SEQ ID NO:47 and the light chain CDR2 and SEQ ID NO:48, a light chain CDR3;
(9) SEQ ID NO:49, heavy chain CDR1, SEQ ID NO:50, a heavy chain CDR2, SEQ ID NO:51, heavy chain CDR3, SEQ ID NO:52, light chain CDR1, SEQ ID NO:53 and the light chain CDR2 and SEQ ID NO:54 light chain CDR3; or (b)
(10) SEQ ID NO:55, heavy chain CDR1, SEQ ID NO:56, heavy chain CDR2, SEQ ID NO:57, a heavy chain CDR3, SEQ ID NO:58, light chain CDR1, SEQ ID NO:59 and light chain CDR2 and SEQ ID NO:60, and a light chain CDR3.
In one embodiment of the invention, the antigen binding region is an scFv or Fab.
In one embodiment of the invention, the extracellular region further comprises a hinge region. In one embodiment of the invention, the hinge region comprises a human CD 8a hinge region. In one embodiment of the invention, the human CD 8a hinge region has the amino acid sequence of SEQ ID NO: 63.
In one embodiment of the invention, the transmembrane region comprises a human CD8 a transmembrane region. In one embodiment of the invention, the human CD8 a transmembrane region has the amino acid sequence of SEQ ID NO: 65.
In one embodiment of the invention, the intracellular region comprises one or more signal transduction regions. In one embodiment of the invention, the intracellular region comprises a human 41BB intracellular region and/or a human cd3ζ intracellular region. In one embodiment of the invention, the human 41BB intracellular region has the amino acid sequence of SEQ ID NO:67 and/or said human cd3ζ intracellular domain has the amino acid sequence shown in SEQ ID NO: 69.
In a further aspect, the invention relates to a polypeptide characterized by comprising a signal peptide and a chimeric antigen receptor according to the invention. In one embodiment of the invention, the signal peptide is a CD8 a signal peptide. In one embodiment of the invention, the CD8 a signal peptide has the amino acid sequence of SEQ ID NO:61, and a sequence of amino acids shown in seq id no.
In a further aspect, the invention relates to a polynucleotide, characterized in that it encodes a chimeric antigen receptor according to the invention or a polypeptide according to the invention.
In a further aspect, the invention relates to a vector, characterized in that it comprises a polynucleotide according to the invention. In one embodiment of the invention, the vector is a cloning vector or an expression vector. In one embodiment of the invention, the vector is a viral vector.
In a further aspect, the invention relates to an engineered immune effector cell, characterized in that it expresses a polypeptide of a chimeric antigen receptor according to the invention, or comprises a polynucleotide encoding a polypeptide of a chimeric antigen receptor or according to the invention. In one embodiment of the invention, the engineered immune effector cell is selected from T lymphocytes, NK cells, pluripotent stem cells or embryonic stem cells, in particular T lymphocytes.
In a further aspect, the invention relates to a method for engineering an immune effector cell, characterized in that the immune effector cell is infected with a vector according to the invention, in particular a viral vector. In one embodiment of the invention, the immune effector cell is a T lymphocyte.
In a further aspect, the invention relates to the use of an antibody or antigen binding fragment, chimeric antigen receptor, polypeptide, polynucleotide, vector, or engineered immune effector cell according to the invention in the manufacture of a medicament for the treatment of cancer. In a further aspect, the invention relates to the use of an antibody or antigen binding fragment, chimeric antigen receptor, polypeptide, polynucleotide, vector, or engineered immune effector cell according to the invention in the manufacture of a medicament for stimulating immune function in a patient having cancer. In yet another aspect, the invention relates to a method of treating cancer comprising administering an antibody or antigen binding fragment, chimeric antigen receptor, polypeptide, polynucleotide, vector, or engineered immune effector cell according to the invention to a patient having the cancer. In a further aspect, the invention relates to a method of stimulating immune function in a patient having cancer comprising administering to the patient an antibody or antigen binding fragment, chimeric antigen receptor, polypeptide, polynucleotide, vector, or engineered immune effector cell according to the invention. In one embodiment of the invention, the cancer is a B cell-related cancer, particularly multiple myeloma. In one embodiment of the invention, the immune function is IFN-gamma secretion and/or CD107a expression
Brief Description of Drawings
FIG. 1 shows the killing of target cells by anti-BCMACAR-T.
FIG. 2 shows IFN-. Gamma.and CD107a expression against BCMACAR-T CD 3-positive cells.
FIG. 3 shows IFN-gamma expression against BCMACAR-T CD 3-positive cells.
FIG. 4 shows CD107a expression against BCMACAR-T CD3 positive cells.
FIG. 5 shows the heavy chain CDR amino acid sequences of an antibody of the present invention (according to the IMGT system).
FIG. 6 shows the light chain CDR amino acid sequences of the antibodies of the present invention (according to the IMGT system).
Detailed Description
Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology, microbiology, cell biology, biochemistry and immunology. These techniques are well within the skill of the art and are well described in the literature.
It should be understood that embodiments of "comprising" as described herein include embodiments that "consist of … …" and/or "consist essentially of.
I. anti-BCMA antibodies
One aspect of the present invention provides anti-BCMA antibodies that specifically bind to BCMA, such as human BCMA, particularly the extracellular domain of human BCMA. In some embodiments, the anti-BCMA antibodies of the invention are monoclonal antibodies.
B Cell Maturation Antigen (BCMA), also known as CD269 or TNFRSF17, is a member of the tumor necrosis factor receptor superfamily. BCMA is expressed almost exclusively in plasma cells and multiple myeloma cells. BCMA may be a suitable tumor antigen target for immunotherapeutic agents against multiple myeloma.
In some embodiments, the anti-BCMA antibody of the present invention is a full length antibody. The terms "full length antibody," "whole antibody," and "whole antibody" are used interchangeably herein to refer to an antibody in substantially complete form as compared to an antibody fragment. The terms "antibody" and "immunoglobulin" are used interchangeably herein. The basic 4-chain antibody unit is a hetero-tetrameric glycoprotein consisting of two identical light (L) chains and two identical heavy (H) chains. The L chains from any vertebrate species can be classified into two distinct types, called kappa (kappa) and lambda (lambda), based on the amino acid sequence of their constant regions. Antibodies can be categorized into five different classes based on the amino acid sequence of the heavy chain constant region: igA, igD, igE, igG and IgM, which have heavy chains called α, δ, ε, γ and μ, respectively. IgG and IgA classes can be further divided into subclasses such as IgG1, igG2 (including IgG2A and IgG 2B), igG3, igG4, igA1, and IgA2. In the case of IgG, the 4-chain unit is typically about 150,000 daltons. Each H chain has a variable domain at the N-terminus (V H) followed by three constant domains per gamma chain (C H). Each L chain has a variable domain at the N-terminus (V L) followed by a constant domain at the other end (C L).
In some embodiments, the anti-BCMA antibodies of the invention are IgG, particularly IgG 1. In some embodiments, the anti-BCMA antibodies of the invention are kappa.
In some embodiments, the anti-BCMA antibodies of the invention are antibody fragments. The term "antibody fragment" comprises a portion of an intact antibody, preferably the antigen binding and/or variable regions of an intact antibody. Examples of antibody fragments include Fab, fab '-SH, F (ab') 2, fv, and scFv fragments; a double body; a linear antibody; a single chain antibody; a single domain antibody; and multispecific antibodies formed from antibody fragments. The term "scFv" is an antibody fragment comprising V H and V L domains linked into a single polypeptide chain. Preferably, the scFv further comprises a polypeptide linker between the V H and V L domains. The term "Fab" consists of the whole L chain as well as the variable domain of the H chain (V H) and the first constant domain (C H 1).
In some embodiments, an anti-BCMA antibody of the present invention is an antigen binding fragment comprising a heavy chain variable domain and a light chain variable domain of a parent antibody, e.g., scFv or Fab. Antibody fragments can be prepared by a variety of techniques, including, but not limited to, proteolytic digestion of intact antibodies, and production by recombinant host cells.
In some embodiments, the anti-BCMA antibodies of the invention are murine, chimeric, humanized, or human.
In a "chimeric" antibody, a portion of the heavy and/or light chain is identical or homologous to a corresponding sequence in an antibody derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the heavy and/or light chain is identical or homologous to a corresponding sequence in an antibody derived from another species or belonging to another antibody class or subclass, so long as they exhibit the desired biological activity. In some embodiments, the chimeric antibodies of the invention comprise a murine variable region and a human constant region.
A "humanized" form of a non-human (e.g., murine) antibody is a chimeric antibody that contains minimal sequences derived from the non-human antibody. In some embodiments, the humanized antibody is a human antibody (recipient antibody) in which CDR (or HVR) residues of the recipient antibody are replaced with CDR (or HVR) residues of a non-human species (donor antibody) such as mouse, rat, rabbit or non-human primate having the desired specificity, affinity, and/or capacity. In some cases, framework ("FR") residues of a human antibody are replaced with corresponding non-human residues. In addition, the humanized antibody may comprise residues that are not present in either the recipient antibody or the donor antibody. These modifications may be made to further refine antibody properties such as binding affinity.
A "human antibody" is an antibody having an amino acid sequence corresponding to that of a human-produced antibody and/or prepared using any technique for preparing a human antibody. This definition of human antibodies specifically excludes humanized antibodies that comprise non-human antigen binding residues. Human antibodies can be produced using a variety of techniques known in the art. Human antibodies can be prepared by administering an immunogen to a transgenic animal that has been modified to produce such antibodies in response to antigen challenge, but whose endogenous loci have been disabled. Human antibodies can also be produced by phage display libraries of human origin. The library of immune sources provides high affinity antibodies to the immunogen without the need to construct hybridomas. Alternatively, the unexposed pool can be cloned to give antibodies to a variety of non-self and self antigens without any immunization.
In some embodiments, an anti-BCMA antibody or antigen binding fragment of the invention comprises the heavy chain CDRs shown in fig. 5. In some embodiments, an anti-BCMA antibody or antigen binding fragment of the invention comprises the light chain CDRs shown in fig. 6.
In some embodiments, the invention encompasses antibodies that bind to the same BCMA epitope as any of the anti-BCMA antibodies described herein. In some embodiments, the invention encompasses antibodies that compete with any of the anti-BCMA antibodies described herein for binding to BCMA. In some embodiments, competition assays can be used to identify antibodies that bind to the same BCMA epitope as any of the anti-BCMA antibodies described herein or antibodies that compete for binding to BCMA with any of the anti-BCMA antibodies described herein. In some embodiments, two antibodies are considered to bind to the same epitope if one blocks the binding of the other to the antigen by 50% or more.
One aspect of the present invention provides one or more polynucleotides characterized by encoding an anti-BCMA antibody according to the present invention. In one aspect, the invention provides a vector, characterized in that it comprises a polynucleotide according to the invention. In one embodiment, the vector is a cloning vector or an expression vector, in particular a plasmid vector. In one aspect the invention provides a host cell characterized by comprising a polynucleotide according to the invention or a vector according to the invention. In one embodiment, the host cell is a prokaryotic cell, in particular an E.coli cell. In another embodiment, the host cell is a eukaryotic cell, particularly a chinese hamster ovary cell. In one aspect, the invention provides a method for producing an anti-BCMA antibody, characterized by culturing a host cell according to the invention under conditions for expression of the antibody.
Chimeric antigen receptor
One aspect of the invention provides a Chimeric Antigen Receptor (CAR) that targets BCMA. Chimeric antigen receptors are generally composed of an extracellular region, a transmembrane region, and an intracellular region. The extracellular region comprises an antigen binding region, and optionally a hinge region. The intracellular region comprises one or more signaling regions, including a costimulatory signaling region. The polypeptide of the chimeric antigen receptor may also comprise a signal peptide when expressed in a cell.
Signal peptides
When expressed in a cell, the polypeptide of the chimeric antigen receptor of the invention may comprise a signal peptide (also referred to as a signal sequence) at the N-terminus of the polypeptide. In general, a signal peptide is a peptide sequence that targets a polypeptide to a desired site in a cell. In some embodiments, the signal peptide targets the polypeptide to the secretory pathway of the cell and will allow the polypeptide to integrate and anchor to the lipid bilayer.
In some embodiments, the signal peptide used in the present invention is derived from CD8 a. In some embodiments, the CD 8a signal peptide comprises SEQ ID NO:61, and a sequence of amino acids. In some embodiments, the CD 8a signal peptide consists of SEQ ID NO: 62.
Antigen binding region
The chimeric antigen receptor of the invention comprises an antigen binding region that targets BCMA. The antigen binding region may be monovalent or multivalent (e.g., bivalent). The antigen binding region may also be monospecific or multispecific (e.g., bispecific). Bispecific may be against BCMA and another antigen, or against two different epitopes of BCMA.
In some embodiments, the antigen binding regions used in the present invention are in the form of an anti-BCMA antibody or antigen binding fragment described above, particularly scFv.
Hinge region
Optionally, the chimeric antigen receptor of the invention comprises a hinge region between the extracellular antigen binding region and the transmembrane region. The hinge region is a segment of amino acids that typically exist between two domains of a protein and can allow flexibility of the protein and movement of the two domains relative to each other.
The hinge region may be the hinge region of a naturally occurring protein or a portion thereof. The hinge region of an antibody (such as IgG, igA, igM, igE or IgD antibodies) can also be used for the chimeric antigen receptor described herein. Non-naturally occurring peptides can also be used as hinge regions for chimeric antigen receptors described herein. In some embodiments, the hinge region is a peptide linker.
In some embodiments, the hinge region used in the present invention is derived from CD8 a. In some embodiments, the CD 8a hinge region comprises SEQ ID NO: 63. In some embodiments, the CD 8a hinge region consists of SEQ ID NO:64, a nucleic acid sequence encoding a polypeptide.
Transmembrane region
The chimeric antibody receptor of the invention comprises a transmembrane region. The transmembrane region may form an alpha helix, a complex of more than one alpha helix, a beta barrel, or any other stable structure capable of crossing a domain cellular phospholipid bilayer. The transmembrane region may be of natural or synthetic origin. The transmembrane region may be derived from the alpha, beta or zeta chain of T cell receptor 、CD28、CD3ε、CD45、CD4、CD5、CD8、CD9、CD16、CD22、CD33、CD37、CD64、CD80、CD86、CD134、CD137、CD154、KIRDS2、OX40、CD2、CD27、LFA-1(CD11a、CD18)、ICOS(CD278)、4-1BB(CD137)、GITR、CD40、BAFFR、HVEM(LIGHTR)、SLAMF7、NKp80(KLRF1)、CD160、CD19、IL-2Rβ、IL-2Rγ、IL-7Ra、ITGA1、VLA1、CD49a、ITGA4、IA4、CD49D、ITGA6、VLA-6、CD49f、ITGAD、CDlld、ITGAE、CD103、ITGAL、CD11a、LFA-1、ITGAM、CDllb、ITGAX、CD11c、ITGB1、CD29、ITGB2、CD18、LFA-1、ITGB7、TNFR2、DNAM1(CD226)、SLAMF4(CD244、2B4)、CD84、CD96(Tactile)、CEACAM1、CRT AM、Ly9(CD229)、CD160(BY55)、PSGL1、CDIOO(SEMA4D)、SLAMF6(NTB-A、Lyl08)、SLAM(SLAMF1、CD150、IPO-3)、BLAME(SLAMF8)、SELPLG(CD162)、LTBR、PAG/Cbp、NKp44、NKp30、NKp46、NKG2D and/or NKG 2C.
In some embodiments, the transmembrane region used in the present invention is derived from CD8 a. In some embodiments, the CD8 a transmembrane region comprises SEQ ID NO: 65. In some embodiments, the CD8 transmembrane region consists of SEQ ID NO: 66.
Intracellular region
The chimeric antigen receptor of the invention comprises an intracellular region. The intracellular region comprises one or more signaling regions, including a costimulatory signaling region.
The intracellular signaling region is responsible for activation of at least one normal effector function of immune effector cells expressing the chimeric antigen receptor. For example, the effector function of a T cell may be a cell lysis activity or a helper activity, including secretion of cytokines. Although the entire intracellular signaling region is generally available, in many cases, the use of an entire strand is not necessary. In the case of using a truncated portion of the intracellular signaling region, such a truncated portion may be used instead of the complete strand as long as it transduces the effector function signal. Thus, an intracellular signaling region includes any truncated form of the intracellular signaling region sufficient to transduce an effector function signal. In some embodiments, the signaling region is derived from cd3ζ, fcrγ (FCER 1G), fcrβ (fcεrib), cd3γ, cd3δ, cd3ε, CD5, CD22, CD79a, CD79b, and CD66d.
In some embodiments, the signaling region used in the present invention is derived from cd3ζ. In some embodiments, the CD3 zeta signaling region comprises SEQ ID NO: 69. In some embodiments, the CD3 zeta signaling region consists of SEQ ID NO: 70.
In some embodiments, the intracellular regions of the chimeric antigen receptor of the invention further comprise one or more costimulatory signaling regions. In addition to stimulation of antigen specific signals, many immune effector cells require co-stimulation to promote cell proliferation, differentiation and survival, as well as to activate the effector functions of the cells. The "costimulatory signaling region" may be the cytoplasmic portion of a costimulatory molecule. The term "costimulatory molecule" refers to a cognate binding partner on an immune cell (such as a T cell) that specifically binds to a costimulatory ligand, thereby mediating a costimulatory response, such as, but not limited to, proliferation and survival, by the immune cell. The costimulatory signaling region may be derived from a B7/CD28 family member (e.g., B7-1/CD80、B7-2/CD86、B7-H1/PD-L 1、B7-H2、B7-H3、B7-H4、B7-H6、B7-H7、BTLA/CD272、CD28、CTLA-4、Gi24/VISTA/B7-H5、ICOS/CD278、PD-1、PD-L2/B7-DC and PDCD 6); TNF superfamily members (e.g., 4-1BB/TNFSF9/CD137, 4-1BB ligand/TNFSF 9, BAFF/BLyS/TNFSF13B, BAFF R/TNFRSF13C, CD/TNFRSF 7, CD27 ligand/TNFSF 7, CD30/TNFRSF8, CD30 ligand/TNFSF 8, CD40/TNFRSF5, CD40/TNFSF5, CD40 ligand/TNFSF 5, DR3/TNFRSF25, GITR/TNFRSF18, GITR ligand/TNFSF 18, HVEM/TNFRSF14, LIGHT/TNFSF14, lymphotoxin-alpha/TNF-beta, OX40/TNFRSF4, OX40 ligand/TNFSF 4, RELT/TNFRSF19L, TACI/TNFRSF13B, TL A/TNFSF15, TNF-alpha and TNF-gamma/TNFSF 1B); SLAM family members (e.g., ,2B4/CD244/SLAMF4、BLAME/SLAMF8、CD2、CD2F-10/SLAMF9、CD48/SLAMF2、CD58/LFA-3、CD84/SLAMF5、CD229/SLAMF3、CRACC/SLAMF7、NTB-A/SLAMF6 and SLAM/CD 150); and any other costimulatory molecules such as CD2, CD7, CD53, CD82/Kai-1, CD90/Thy1, CD96, CD160, CD200, CD300a/LMIR1, HLA class I, HLA-DR, ikaros, integrin alpha 4/CD49d, integrin alpha 4 beta 1, integrin α4β7/LPAM-1、LAG-3、TCL1A、TCL1B、CRTAM、DAP12、Dectin-1/CLEC7A、DPPIV/CD26、EphB6、TIM-1/KIM-1/HAVCR、TIM-4、TSLP、TSLP R、 lymphocyte function-associated antigen-1 (LFA-1) and NKG2C.
In some embodiments, the costimulatory signaling region used in the present invention is derived from 4-1BB. In some embodiments, the 4-1BB costimulatory signaling region comprises the sequence of SEQ ID NO: 67. In some embodiments, the 4-1BB costimulatory signaling region consists of the sequence set forth in SEQ ID NO:68, and a nucleic acid sequence encoding the same.
In some embodiments, the intracellular region of a chimeric antigen receptor of the invention comprises the 4-1BB costimulatory signaling region described above and the CD3 zeta signaling region described above linked in an N-terminal to C-terminal direction.
In one aspect the invention provides a polynucleotide which encodes a chimeric antigen receptor or polypeptide according to the invention. In one aspect, the invention provides a vector, characterized in that it comprises a polynucleotide according to the invention. In one embodiment, the vector is a cloning vector or an expression vector. In one embodiment, the vector is a viral vector.
Engineered immune effector cells
One aspect of the invention provides cells, such as immune effector cells, engineered to contain or express the BCMA-targeted chimeric antigen receptor of the invention. In some embodiments, the immune effector cells are T cells, NK cells, peripheral Blood Mononuclear Cells (PBMCs), hematopoietic stem cells, pluripotent stem cells, or embryonic stem cell culture differentiated cells (e.g., immune cells). In some embodiments, the immune effector cell is autologous. In some embodiments, the immune effector cell is allogeneic.
Carrier body
One aspect of the present invention provides a vector for cloning and expressing the BCMA-targeted chimeric antigen receptor of the present invention. In some embodiments, the vector is suitable for replication and integration in eukaryotic cells (such as mammalian cells). In some embodiments, the vector is a viral vector. Examples of viral vectors include, but are not limited to, adenovirus vectors, adeno-associated virus vectors, lentiviral vectors, retrovirus vectors, vaccinia vectors, herpes simplex virus vectors, and derivatives thereof.
Many virus-based systems have been developed for gene transfer to mammalian cells. In some embodiments, lentiviral vectors are used. In some embodiments, self-inactivating lentiviral vectors are used. For example, self-inactivating lentiviral vectors carrying chimeric antigen receptor coding sequences may be packaged using protocols known in the art. The resulting lentiviral vector may be used to transduce mammalian cells (such as primary human T cells) using methods known in the art. Lentiviral derived vectors are suitable tools for achieving long term gene transfer because they allow long term, stable integration of transgenes and their propagation in daughter cells. Lentiviral vectors are also low immunogenic and can transduce non-proliferating cells.
Immune effector cells
An "immune effector cell" is an immune cell that can perform an immune effector function. In some embodiments, the immune effector cells express at least fcyriii and perform ADCC effector function. Examples of immune effector cells that mediate ADCC include Peripheral Blood Mononuclear Cells (PBMCs), natural Killer (NK) cells, monocytes, cytotoxic T cells, neutrophils, and eosinophils.
In some embodiments, the immune effector cell is a T cell. In some embodiments, the T cell is CD4+/CD8-, CD4-/CD8+, CD4+/CD8+, CD4-/CD8-, or a combination thereof. In some embodiments, the T cell produces IL-2, IFN, and/or TNF when expressing the chimeric antigen receptor and binding to the target cell. In some embodiments, cd8+ T cells lyse antigen-specific target cells when expressing chimeric antigen receptors and binding to the target cells.
Engineered immune effector cells are prepared by introducing chimeric antigen receptors into immune effector cells, such as T cells. In some embodiments, the chimeric antigen receptor is introduced into the immune effector cell by transfecting a nucleic acid or vector comprising a sequence encoding the chimeric antigen receptor. In some embodiments, the chimeric antigen receptor is introduced into the immune effector cell by inserting the protein into the cell membrane while passing the cell through a microfluidic system.
Methods for introducing nucleic acids or vectors into mammalian cells are known in the art. The vector may be transferred into immune effector cells by physical, chemical or biological means. Physical methods for introducing the vector into immune effector cells include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Chemical means for introducing nucleic acids or vectors into immune effector cells include colloidal dispersions such as macromolecular complexes, nanocapsules, microspheres, beads and lipid-based systems (including oil-in-water emulsions, micelles, mixed micelles and liposomes). An exemplary colloidal system for use as an in vitro delivery vehicle is a liposome (e.g., an artificial membrane vesicle). Biological methods for introducing nucleic acids or vectors into immune effector cells include the use of DNA and RNA vectors. Viral vectors have become the most widely used method for inserting genes into mammalian, e.g., human, cells.
In some embodiments, the transduced or transfected immune effector cells are propagated ex vivo after introduction of the nucleic acid or vector. In some embodiments, the transduced or transfected immune effector cells are further evaluated or screened to select for engineered immune effector cells.
T cell origin
Prior to expansion and genetic modification of T cells, a source of T cells is obtained from an individual. T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from an infection site, ascites, pleural effusion, spleen tissue, and tumors.
Activation and expansion of T cells
Whether before or after genetic modification of T cells using the chimeric antigen receptors described herein, T cells can generally be activated and expanded using methods known in the art.
In general, T cells can be expanded by contacting a surface to which are attached reagents that stimulate a CD3/TCR complex-associated signal and ligands that stimulate co-stimulatory molecules on the surface of the T cells. In particular, the anti-CD 3 antibody or antigen-binding fragment thereof or the anti-CD 2 antibody immobilized on a surface may be contacted, or by contacting a protein kinase C activator (e.g., bryostatin) in combination with a calcium ionophore. For co-stimulation of helper molecules on the T cell surface, ligands that bind to the helper molecules are used. For example, a population of T cells may be contacted with an anti-CD 3 antibody and an anti-CD 28 antibody under conditions suitable to stimulate T cell proliferation. To stimulate proliferation of cd4+ T cells or cd8+ T cells, anti-CD 3 antibodies and anti-CD 28 antibodies are used.
In some embodiments, the primary stimulation signal and the co-stimulation signal of the T cells may be provided by different protocols. For example, the reagents providing each signal may be in solution or bound to a surface. When the agent binds to a surface, it can bind to the same surface (i.e., in "cis" form) or a separate surface (i.e., in "trans" form). Or one reagent may be bound to the surface while the other reagent is in solution. In one embodiment, the agent that provides the co-stimulatory signal binds to the cell surface, while the agent that provides the primary activation signal is in solution or bound to the surface. In certain embodiments, both reagents may be in solution. In another embodiment, the agent may be in a soluble form and then crosslinked to the surface.
In some embodiments, T cells are combined with reagent coated beads, followed by separation of the beads and cells, and then culturing the cells. In an alternative embodiment, the reagent coated beads and cells are not separated but are cultured together prior to culturing. In another embodiment, the beads and cells are first concentrated by applying a force (such as a magnetic force) such that the attachment of cell surface markers is increased, thereby inducing cell stimulation.
Suitable conditions for T cell culture include suitable media (e.g., minimal essential media or RPMI media 1640) that may contain factors necessary for proliferation and survival, including serum (e.g., fetal bovine serum or human serum), interleukin-2 (IL-2), insulin, IFN-gamma, IL-4, IL-7, GM-CSF, IL-10, IL-12, IL-15, TGF beta, and TNF-alpha, or any other additives known to the skilled artisan for cell growth. Other additives for cell growth include, but are not limited to, surfactants, human plasma protein powder (plasmanate), and reducing agents such as N-acetylcysteine and 2-mercaptoethanol. The culture medium may include RPMI 1640, AIM-V, DMEM, MEM, alpha-MEM, F-12, X-Vivo15 and X-Vivo20 supplemented with amino acids, sodium pyruvate and vitamins, which are serum-free or supplemented with an appropriate amount of serum (or plasma) or a defined set of hormones, and/or an amount of cytokines sufficient for T cell growth and expansion. Antibiotics (e.g., penicillin and streptomycin) are included only in the experimental cultures, and not in the cell cultures to be infused into the subject. The cells are maintained under conditions required to support growth, such as an appropriate temperature (e.g., 37 ℃) and atmosphere (e.g., air plus 5% CO 2). T cells that have been exposed to different stimulation times may exhibit different characteristics. For example, typical peripheral blood mononuclear cell products of blood or apheresis have a helper T cell population (TH, cd4+), which is larger than a cytotoxic or suppressor T cell population (TC, CD 8). Ex vivo expansion of T cells by stimulation of CD3 and CD28 receptors results in a T cell population consisting essentially of TH cells prior to about day 8-9, whereas after about day 8-9, the T cell population comprises a progressively increasing population of TC cells. Thus, depending on the therapeutic purpose, it may be advantageous to infuse the subject with a T cell population comprising predominantly TH cells. Similarly, if an antigen-specific subset of TC cells has been isolated, it may be beneficial to expand that subset to a greater extent.
Furthermore, other phenotypic markers besides the CD4 and CD8 markers are also significantly different, but to a large extent reproducible during cell expansion. Thus, this reproducibility enables the tailoring of the activated T cell product for a specific purpose.
As used herein, the expressions "cell", "cell line" and "cell culture" are used interchangeably and all of these designations include progeny. It is understood that all progeny may not be exactly identical in DNA content, due to deliberate or inadvertent mutation. Including variant progeny that have the same function or biological activity as the cells selected in the initially transformed cells.
The terms "host cell", "host cell line" and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include "transformants" and "transformed cells" which include primary transformed cells and progeny derived therefrom, regardless of the number of passages. The progeny may not be exactly identical in nucleic acid content to the parent cell, but may contain mutations. Included herein are mutant progeny that have the same function or biological activity as the cell selected or selected in the originally transformed cell.
Diagnostic and detection methods and uses
One aspect of the invention provides a method for detecting BCMA in a biological sample. The term "detection" encompasses quantitative or qualitative detection. One aspect of the invention provides a method for diagnosing cancer. In one embodiment, the cancer includes, but is not limited to, B cell related cancers, particularly multiple myeloma. In one embodiment, the cancer includes, but is not limited to, hematological malignancies and solid tumors, particularly multiple myeloma.
In some embodiments, the method comprises contacting the biological sample with an anti-BCMA antibody described herein under conditions that allow the anti-BCMA antibody to bind BCMA, and detecting whether a complex is formed between the anti-BCMA antibody and BCMA, wherein the formation of the complex indicates the presence of BCMA in the biological sample or that the subject producing the biological sample has cancer. Such methods may be in vitro or in vivo.
In one embodiment, a labeled anti-BCMA antibody or antigen binding fragment is provided. Labels include, but are not limited to, directly detected labels or moieties (such as fluorescent labels, chromophore labels, electron density labels, chemiluminescent labels, and radioactive labels), and indirectly detected labels or moieties (e.g., by enzymatic reactions or molecular interactions, such as enzymes with substrates, receptors, and ligands).
V. methods of treatment and uses
One aspect of the invention provides methods for treating cancer, or for stimulating immune function in a patient having cancer. In one embodiment, the method comprises administering one or more antibodies or antigen binding fragments of the invention. In one embodiment, the method comprises administering a polynucleotide encoding one or more antibodies or antigen binding fragments of the invention. In one embodiment, the method comprises engineering an immune effector cell to express one or more chimeric antigen receptors of the invention. In one embodiment, the method comprises administering one or more engineered immune effector cells of the invention. In one embodiment, the method is cellular immunotherapy.
In one embodiment, the cancer includes, but is not limited to, B cell related cancers, particularly multiple myeloma. In one embodiment, the cancer includes, but is not limited to, hematological malignancies and solid tumors, particularly multiple myeloma.
Administration may be by any convenient means, including by injection, inhalation, infusion, implantation or implantation. Administration may be intra-arterial, subcutaneous, intradermal, intratumoral, intranodal, intramedullary, intramuscular, intravenous or intraperitoneal. In some embodiments, the administration is systemic or local. Infusion techniques for immunotherapy are known in the art.
The dosage and concentration of the agents of the present invention may vary depending on the particular application. Determination of the appropriate dosage is well within the skill of the ordinarily skilled artisan.
In some embodiments, the pharmaceutical composition is administered in a single administration. In some embodiments, the pharmaceutical composition is administered multiple times, such as2, 3, 4, 5,6, or more times. In some embodiments, the pharmaceutical composition is administered once a week, once a week 3, once a week 4, once a month 2, once a month 3, once a month 4, once a month 6, or once a year.
In some embodiments, the dose may be administered by one or more separate administrations or by continuous infusion. In some embodiments, the pharmaceutical compositions are administered separately, such as in 2, 3, 4, 5, or more administrations. In some embodiments, the divided doses are administered within about one week. In some embodiments, the dose is aliquoted.
The progress of the treatment can be readily monitored by conventional techniques and assays. The optimal dosage and treatment regimen for a particular patient can be readily determined by one skilled in the medical arts by monitoring the patient's signs and adjusting the treatment accordingly.
In some embodiments, the amount of the pharmaceutical composition is effective to elicit an objective clinical response in the individual. In some embodiments, the amount of the pharmaceutical composition is effective to cause remission (partial or complete) of the disease in the subject. In some embodiments, the amount of the pharmaceutical composition is effective to prevent recurrence of cancer or disease progression in the subject. In some embodiments, the amount of the pharmaceutical composition is effective to extend survival of the individual (such as disease-free survival). In some embodiments, the pharmaceutical composition is effective to improve the quality of life of the individual. In some embodiments, the amount of the pharmaceutical composition is effective to inhibit the growth of or reduce the size of a solid or lymphoid tumor. In some embodiments, the amount of the pharmaceutical composition is effective to inhibit tumor metastasis in the subject.
VI pharmaceutical composition
One aspect of the invention provides a pharmaceutical composition comprising one or more antibodies or antigen binding fragments, chimeric antigen receptors or engineered immune effector cells of the invention, and one or more pharmaceutically acceptable carriers.
The pharmaceutical compositions may be prepared in the form of lyophilized formulations or aqueous solutions by mixing the active agent of the desired purity with an optional pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are nontoxic to recipients at the dosages and concentrations employed, and include buffers, antioxidants, preservatives, isotonic agents, stabilizers, surfactants, and the like.
In order for pharmaceutical compositions to be useful for in vivo administration, they must be sterile. The pharmaceutical composition may be rendered sterile by filtration through a sterile filtration membrane.
The pharmaceutical compositions may contain more than one active agent as required for the particular indication to be treated, preferably those having complementary activities that do not adversely affect each other. Alternatively or in addition, the pharmaceutical composition may further comprise a cytotoxic agent, a chemotherapeutic agent, a cytokine, an immunosuppressant, or a growth inhibitory agent. Such molecules are suitably present in combination in an amount effective for the intended purpose.
VII kits and articles of manufacture
One aspect of the invention provides kits and articles of manufacture comprising one or more antibodies or antigen binding fragments, chimeric antigen receptors, or engineered immune effector cells of the invention.
The kit or article of manufacture may comprise a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, and the like. The container may be formed from a variety of materials, such as glass or plastic. The label or package insert includes instructions for use, meaning instructions typically included in commercial packages of products, containing information about the indication, usage, dosage, administration, contraindications and/or warnings of use of such products. In addition, the kit or article of manufacture may also include other materials, including diluents, filters, needles and syringes, as desired from a commercial and user standpoint.
The following examples are provided by way of illustration and not by way of limitation as a mere example of the invention and should not be construed as limiting the invention in any way.
Examples
Example 1: construction of expression vector of fusion protein (BCMA-huIgG 1 Fc) of recombinant human B Cell Maturation Antigen (BCMA) extracellular domain and human IgG1 Fc region and eukaryotic expression
1. Gene synthesis of recombinant BCMA extracellular domain and expression vector construction of BCMA-huIgG1 Fc fusion protein
A gene sequence encoding the 1 st methionine to 54 th alanine interval of human B Cell Maturation Antigen (BCMA) (NCBI access No. np_ 001183.2) encoding the amino acid sequence of SEQ ID NO:71, and a nucleotide sequence shown in seq id no. The gene sequence of the 99 th glutamic acid to 330 th lysine amino acid interval of the human IgG1 heavy chain constant region (UniProtKB/Swiss-Prot access No. P01857.1) encoding the sequence of SEQ ID NO:73, and a nucleotide sequence shown in seq id no. Primers containing the fusion protein signal peptide (with the amino acid sequence shown in SEQ ID NO: 75) gene sequence were chemically synthesized for expression vector construction. The BCMA gene fragment was spliced with the human IgG1 Fc gene fragment by molecular cloning. The splice products were cloned into pcdna3.1 (Thermo) using TaKaRa seamless cloning kit.
2. Expression and purification of recombinant BCMA-huIgG1 Fc fusion proteins
After transfection of 293T cells (ATCC) with this expression vector for 5 days, the culture supernatant was collected and the recombinant BCMA-huIgG1 Fc fusion protein was purified with AKTA explorer 100 (GE). Due to glycosylation modification and other reasons, the recombinant BCMA-huIgG1 Fc fusion protein is subjected to reducing SDS-PAGE electrophoresis and then is stained with Coomassie brilliant blue to show that the recombinant BCMA-huIgG1 Fc fusion protein is about 40 kilodaltons in size.
Example 2: construction of chimeric antibody expression vector of anti-human BCMA murine antibody C11D5.3 variable region and human IgG 1/kappa constant region and eukaryotic expression
1.Acquisition of variable region Gene of murine antibody of C11D5.3 and construction of expression vector of murine-human chimeric antibody of C11D5.3
The c11d5.3 antibody light chain variable region gene is synthesized by a chemical synthesis method, and the c11d5.3 antibody light chain variable region has a sequence of SEQ ID NO:77, and the c11d5.3 antibody heavy chain variable region has the amino acid sequence shown in SEQ ID NO:79, and a sequence of amino acids shown in seq id no. The heavy chain variable region gene is used as a template, the heavy chain variable region fragment is amplified by PCR, and the amplified product is cloned into pFUSEss-CHIg-hG1 (invivogen) containing signal peptide and human IgG1 heavy chain constant region gene by using TaKaRa seamless cloning kit. The light chain variable region gene is used as a template, the light chain variable region fragment is amplified by PCR, and the amplified product is cloned into pFUSE2ss-CLIg-hK (invivogen) containing signal peptide and human kappa light chain constant region gene by using a TaKaRa seamless cloning kit.
Expression and purification of chimeric antibodies of C11D5.3
After 5 days of cotransfection of 293T cells (ATCC) in this double plasmid 1:1 ratio, the culture supernatants were collected and the C11D5.3 chimeric antibody was purified with AKTAexplorer (GE). The C11D5.3 chimeric antibody was electrophoresed on non-reducing SDS-PAGE and stained with Coomassie blue to show that it was approximately 150 kDalton in size.
Example 3: ELISA detection of binding of recombinant human BCMA to C11D5.3 chimeric antibody
The binding of recombinant human BCMA to the c11d5.3 chimeric antibody was detected using an enzyme-linked immunosorbent assay (ELISA) which was performed as follows: the BCMA-huIgG1 Fc fusion protein prepared above was added to the microwell plate at 100 ng/well and coated overnight at 4 ℃. The cells were washed three times with PBS, 1% BSA/PBS was added thereto, 200 uL/well, and the cells were blocked at 37℃for 1 hour. 100 ng/well of C11D5.3 chimeric antibody was added and bound for 1 hour at 37 ℃. PBST was washed three times and HRP-goat anti-human IgG (Fab-specific) was added to bind for 1 hour at 37 ℃. PBST was washed three times, 100 uL/well TMB developing solution was added, developed at 37℃for 10 minutes, 100 uL/well ELISA stop solution was added, and the OD450 value was read by an ELISA reader. The OD450 value reflects the binding of the c11d5.3 chimeric antibody to recombinant human BCMA.
Example 4: preparation of anti-human BCMA fully human antibody
1. Construction of high-capacity human-derived Natural antibody phage library
1.1 Collection of human peripheral blood cell resources and cDNA acquisition
Human peripheral blood cells from different individuals who volunteered to donate were collected, and total RNA was extracted from the above peripheral blood cells with Trizol RNA extraction kit (Invitrogen).
First strand cDNA was synthesized using SuperScriptTM IV First-STRAND SYNTHESIS SYSTEM (Invitrogen) kit using the RNA as a template.
1.2 Antibody Gene amplification
The cDNA was used as a template to amplify the heavy chain variable domain and CH1 constant domain genes by PCR using the heavy chain variable domain upstream primer (containing SfiI cleavage site) and the heavy chain constant domain CH1 downstream primer (containing SfiI cleavage site), and the variable domain and constant domain genes of the kappa He Lam chain by PCR using the variable domain upstream primer (containing NheI cleavage site) and the constant domain downstream primer (containing SalI cleavage site) of the kappa He Lam chain. Into a 50uL reaction system, 25uL phusion master mix (Thermo), 2.5uL (25 pmol) of the upstream primer, 2.5uL (25 pmol) of the downstream primer, 1.5uL DMSO,0.5uL cDNA and 18uL of ddH 2 O were added, respectively. The PCR reaction was performed according to the following procedure: after 1 minute of pre-denaturation at 98 ℃, the mixture enters a temperature cycle, denaturation at 98 ℃ for 30 seconds, annealing at 58 ℃ for 30 seconds, extension at 72 ℃ for 1.5 minutes, and cycle 32 times, and final extension at 72 ℃ for 10 minutes. The amplified heavy chain variable domain and CH1 constant domain genes and variable domain and constant domain genes of kappa or lambda chains were recovered using a DNA gel recovery kit (Invitrogen).
1.3 Construction of a humanized Natural antibody library
The heavy chain variable domain and CH1 constant domain genes were digested with SfiI and the kappa He Lam delta chain variable domain and constant domain genes were digested with NheI (NEB) and SalI (NEB). The digested gene fragment was recovered using a DNA gel recovery kit.
Preparation of a sufficient amount of engineered pComb3XTT phagemids (Scripps institute, usa), wherein: the 5 'side of the light chain variable domain and constant domain gene sequences deleted the SfiI cleavage site, the NheI cleavage site was introduced, and the 3' side of the light chain variable domain and constant domain gene sequences introduced the SalI cleavage site. The 5' side of the heavy chain variable domain and constant domain gene sequences was introduced with SfiI cleavage sites. The amber stop codon of the original phagemid was deleted.
The vector was digested with NheI and SalI, and the digested vector was recovered using a DNA gel recovery kit. The digested phagemid vector was subjected to overnight ligation with the variable and constant domain genes of the digested kappa He Lam-d strand using DNA T4 ligase (NEB). The ligation products were purified using plasmid DNA purification kit (Invitrogen). The recombinant light chain-containing vector was introduced into TG1 competence (Lucigen) by electrotransformation, the recombinant light chain-containing phagemid vector was amplified by culture of escherichia coli, and a sufficient amount of the recombinant light chain-containing phagemid vector was extracted using plasmid extraction kit (Invitrogen). And (3) utilizing SfiI enzyme to cleave the phagemid vector containing the recombinant light chain, and utilizing a DNA gel recovery kit to recover the enzyme-cleaved phagemid vector. The digested phagemid vector was subjected to overnight ligation with the digested heavy chain variable domain and CH1 constant domain genes using DNAT4 ligase. The ligation product was purified using a plasmid DNA purification kit. Vectors containing recombinant light and heavy chains were electrotransformed into SS320 competence using an electrotransformation apparatus (Bio-Rad). 10ul of the electrotransformed bacteria were removed, counted and the phage antibody library size counted by reasonable dilution and streaking on plates containing tetracycline and ampicillin. The remaining electrotransformed bacteria were added to a 2XYT medium containing 5ug/mL tetracycline, 100ug/mL ampicillin and 2% glucose and incubated in a heated incubator. After the completion of the culture, the culture was centrifuged at 4000G for 10 minutes at4℃to supplement the precipitant with an appropriate amount of glycerol and stored at-80℃as an antibody strain library. A large capacity human natural antibody phage library is obtained through multiple electrotransformation accumulation.
2. Screening and identification of antibody phage libraries
2.1 Biotinylation of BCMA-huIgG1 Fc fusion proteins
BCMA-huIgG1 Fc fusion proteins were randomly biotinylated using standard procedures provided by EZ-Link Sulfo-NHS-LC-Biotin (Thermo). ELISA was used to verify the binding activity of biotinylated BCMA-huIgG1 Fc fusion protein to the C11D5.3 chimeric antibody.
2.2 Biopanning
The antibody which is combined with the BCMA-huIgG1 Fc fusion protein (especially the extracellular domain of human B Cell Maturation Antigen (BCMA)) is obtained by using the BCMA-huIgG1 Fc fusion protein as a target protein and using biopanning to carry out biopanning on the human natural antibody library. The antibody strain library was resuscitated and grown to log phase, the antibody library was rescued using M13KO7 helper phage (NEB), centrifuged, resuspended in 2XYT medium containing ampicillin and kanamycin, and amplified overnight at 30 ℃. PEG/NaCl precipitation phage, using glycerol/PBST dissolved phage precipitation to obtain natural library phage suspension. Casein (Thermo) blocked phage input Casein blocked biotinylated PD1-huIgG1 Fc fusion protein (PD 1 part contains the 25 th to 167 th amino acid region of NCBI access No. NP-005009.2, as shown in SEQ ID NO:81, the huIgG1 Fc part differs from the huIgG1 Fc part used to construct BCMA-huIgG1 Fc fusion protein only in that it does not contain glutamic acid 99 th) and casein blocked Dynabeads M-270 streptavidin (Thermo) co-incubation system, and the supernatant phage suspension was collected. Further, the collected phage suspension was put into a casein blocked biotinylated BCMA-huIgG1 Fc fusion protein and casein blocked Dynabeads M-270 streptavidin co-incubation system and the beads were washed with PBST to remove phages that could not bind to BCMA-huIgG1 Fc fusion protein. Phage bound to the magnetic beads were eluted with 100mM triethylamine (Thermo) and then neutralized with 1M Tris-HCl (pH-6.4). 10ul of eluted phage solution was left for determination of total phage output, the remaining phage solution was used to infect logarithmically growing SS320, and after overnight amplification was considered as antibody library for the next round of panning. Biopanning was performed three times together, with BCMA-huIgG1 Fc antigen concentration starting at 100nM and screening antigen concentration decreasing at 3-fold gradient per round.
2.3 Screening of BCMA ectodomain specific binding clones
The antibody library obtained after the completion of the third biopanning was diluted and plated on plates containing tetracycline and ampicillin to obtain a monoclonal, and the monoclonal was selected and cultured overnight in a deep well plate. The deep-hole plate is repeatedly frozen and thawed three times by using a refrigerator at the temperature of-20 ℃ in the next day, and the supernatant is used for the subsequent ELISA reaction. ELISA reactions were screened for positive clones using goat anti-human IgG (Fab-specific) overnight coating, followed by addition of the centrifugal supernatant, BCMA-huIgG1 Fc fusion protein and streptavidin-HRP. This screening step was repeated twice for independent experiments to ensure data accuracy.
To exclude clones binding to the huIgG1 Fc region, the following ELISA reactions were performed: screening of huIgG1 Fc region binding positive clones was performed using goat anti-human IgG (Fab specific) overnight coating, followed by addition of the centrifugal supernatant, PD1-huIgG1 Fc fusion protein and streptavidin-HRP. This screening step was repeated twice for independent experiments to ensure data accuracy.
BCMA ectodomain specific binding clones were selected from the two above steps.
2.4 Affinity ranking Using dissociation Rate constant k off
The freeze-thaw supernatants of the BCMA extracellular domain specific binding clones screened in the above step were analyzed using an Octet K2 (ForteBio) molecular interaction analyzer. Biotinylated BCMA-huIgG1 Fc fusion proteins were immobilized on SA probes (ForteBio) and affinity assays were performed with freeze-thaw supernatants as analytes. In the analysis step, only the dissociation curve is selected to calculate the k off value, and the k off value is used as a reference basis to order from small to large. Preferably, clones k off, such as clones HK10, HA08, HD10, HA05, and HD07, are ranked first five.
Example 5: preparation of anti-human BCMA murine antibody
1. Immunization of animals
2Mg/mL BCMA-huIgG1 Fc fusion protein was mixed and emulsified as antigen with an equal volume of complete Freund's adjuvant (Sigma-Aldrich) and 56 week old female Balb/c mice (Shanghai Nannon model animal research center) were immunized subcutaneously. After the primary immunization, booster immunization was performed every ten days, four subcutaneous immunizations were performed in total, and spleen impact immunization was performed directly with BCMA-huIgG1 Fc fusion protein at the fifth immunization.
2. Serum titer detection
Blood was collected from the anterior-caudal vein after each boost for 50uL, and the cells were removed by centrifugation, leaving serum. ELISA microwells were added with 50ng of recombinant BCMA per well and coated overnight at 4 ℃. The cells were washed three times with PBS, 1% BSA/PBS was added thereto, 200 uL/well, and the cells were blocked at 37℃for 1 hour. The mouse serum was added in gradient dilution and combined at 37℃for 1 hour. PBST was washed three times, and HRP-goat anti-mouse IgG was added for binding for 1 hour at 37 ℃. PBST was washed three times, 100 uL/well TMB developing solution was added, developed at 37℃for 10 minutes, 100 uL/well ELISA stop solution was added, and the OD450 value was read by an ELISA reader.
3. Construction of an immune library
3.1 Total cDNA acquisition of mouse spleen cells
The BCMA-huIgG1 Fc fusion protein was directly used for the intraperitoneal injection for impact immunization, and four days later, mice were sacrificed and spleens were taken. The whole spleen was ground with a cell screen (BD) to obtain spleen cells. After washing twice with PBS, 1000g was centrifuged for 10 minutes to obtain spleen cells. Total RNA was extracted using Trizol RNA extraction kit.
First strand cDNA was synthesized using the SuperScript TM IV First-STRAND SYNTHESIS SYSTEM kit using the RNA as template.
3.2 Antibody Gene amplification and light-heavy chain splicing
The cDNA was used as a template to PCR amplify the heavy chain variable domain gene using the heavy chain variable domain upstream primer and downstream primer (VH-F, VH-R), and the light chain variable domain upstream primer and downstream primer (VK-F, VK-R) to PCR amplify the kappa chain variable domain gene. Into a 50uL reaction system, 25uL phusion master mix uL (25 pmol) of the upstream primer, 2.5uL (25 pmol) of the downstream primer, 1.5uL DMSO,0.5uL cDNA and 18uL of ddH 2 O were added, respectively. The PCR reaction was performed according to the following procedure: after 1 minute of pre-denaturation at 98 ℃, the mixture enters a temperature cycle, denaturation at 98 ℃ for 30 seconds, annealing at 58 ℃ for 30 seconds, extension at 72 ℃ for 1 minute, 30 times of cycle and final extension at 72 ℃ for 10 minutes.
The amplified VH gene and VK gene were recovered using a DNA gel recovery kit. Equal amounts of VH gene and VK gene were mixed and used as templates, and scFv genes were amplified by overlap PCR using the upstream primer scFv-F and the downstream primer scFv-R. Into a 50uL reaction system, 25uL phusion master mix uL (25 pmol) of the upstream primer, 2.5uL (25 pmol) of the downstream primer, 1.5uL DMSO,0.5uL cDNA and 18uL of ddH 2 O were added, respectively. The PCR reaction was performed according to the following procedure: after 1 minute of pre-denaturation at 98 ℃, the mixture enters a temperature cycle, denaturation at 98 ℃ for 30 seconds, annealing at 58 ℃ for 30 seconds, extension at 72 ℃ for 1 minute, 30 times of cycle and final extension at 72 ℃ for 10 minutes.
And (3) recovering the amplified scFv gene fragment by using a DNA gel recovery kit.
3.3 Construction of immune library
The scFv gene fragment and pcomb XTT vector (Scripps institute in the United states) were digested with SfiI DNA endonuclease, respectively. Into the 50uL reaction system, sfiI 2uL,10 Xbuffer 5uL, DNA 3ug and ddH 2 O were added to 50uL, respectively. After thoroughly mixing, incubation was carried out at 50℃for 3 hours.
The digested scFv gene fragment and pcomb X vector were recovered using a DNA gel recovery kit. The digested scFv gene fragment was cyclized using T4 ligase and the digested pcomb X vector. In a 50uL reaction system, 1uL of T4 ligase, 5uL of 10 Xbuffer, 100ng of scFv gene, 500ng of pComb3X vector and ddH 2 O to 50uL were added, respectively. After thoroughly mixing, incubation was carried out at 4℃for 16 hours. A small amount of the product was taken and the ligation efficiency was verified by agarose gel electrophoresis.
10UL of the ligation cyclized product was added to the homemade TGI electrotransformation competence and then subjected to electrotransformation using an electrotransformation apparatus. 10ul of the electrotransformed bacteria were removed, counted and the phage antibody library size counted by reasonable dilution and streaking on ampicillin-containing plates. The remaining electrotransformed bacteria were added to 2XYT medium containing 100ug/mL ampicillin and 2% glucose and incubated in a heated incubator. After the completion of the culture, the culture was centrifuged at 4000G for 10 minutes at 4℃to supplement the precipitant with an appropriate amount of glycerol and stored at-80℃as an antibody strain library. scFv immune libraries were obtained by multiple electrotransformation accumulation.
4. Screening and identification of murine immune antibody phage library
4.1 Biotinylation of BCMA-huIgG1 Fc fusion proteins
BCMA-huIgG1 Fc fusion proteins were randomly biotinylated using standard procedures provided by EZ-Link Sulfo-NHS-LC-Biotin. ELISA was used to verify the binding activity of biotinylated BCMA-huIgG1 Fc fusion protein to the C11D5.3 chimeric antibody.
4.2 Biopanning
The antibody binding to the BCMA-huIgG1 Fc fusion protein (especially the extracellular domain of human B Cell Maturation Antigen (BCMA)) is obtained by biopanning the murine immune antibody library by using the BCMA-huIgG1 Fc fusion protein as a target protein and applying biopanning. The antibody strain library was resuscitated and grown to log phase, the antibody library was rescued using M13KO7 helper phage, centrifuged, resuspended in 2XYT medium containing ampicillin and kanamycin and amplified overnight at 30 ℃. PEG/NaCl precipitation phage, using glycerol/PBST dissolved phage precipitation to obtain immune library phage suspension. The casein blocked phage were placed into a casein blocked biotinylated PD1-huIgG1 Fc fusion protein and casein blocked Dynabeads M-270 streptavidin co-incubation system and the supernatant phage suspension was collected. Further, the collected phage suspension was put into a casein blocked biotinylated BCMA-huIgG1 Fc fusion protein and casein blocked Dynabeads M-270 streptavidin co-incubation system and the beads were washed with PBST to remove phages that could not bind to BCMA-huIgG1 Fc fusion protein. Phage bound to the beads were eluted with 100mM triethylamine and then neutralized with 1M Tris-HCl (pH=6.4). 10ul of eluted phage solution was left for determination of total phage output, the remaining phage solution was used to infect log-growing TG1, and after overnight amplification was considered as antibody library for the next round of panning. Biopanning was performed in two rounds with BCMA-huIgG1 Fc antigen concentrations of 50nM and 5nM, respectively.
4.3 Screening of extracellular Domain-specific binding clones of BCMA
The antibody library obtained after the second round of biopanning was diluted and spread on ampicillin-containing plates to obtain monoclonal antibodies, and the monoclonal antibodies were selected and cultured overnight in deep well plates. The deep-hole plate is repeatedly frozen and thawed three times by using a refrigerator at the temperature of-20 ℃ in the next day, and the supernatant is used for the subsequent ELISA reaction. ELISA reactions were screened for positive clones using goat anti-human IgG (Fab-specific) overnight coating, with sequential addition of supernatant, biotinylated BCMA-huIgG1 Fc fusion protein and streptavidin-HRP. This screening step was repeated twice for independent experiments to ensure data accuracy.
To exclude clones binding to the huIgG1 Fc region, the following ELISA reactions were performed. Screening of huIgG1 region binding positive clones was performed using goat anti-human IgG (Fab specific) overnight coating, sequentially adding supernatant, biotinylated PD1-huIgG1 Fc fusion protein and streptavidin-HRP. This screening step was repeated twice for independent experiments to ensure data accuracy.
BCMA ectodomain specific binding clones were selected from the two above steps.
4.4 Affinity ranking Using dissociation Rate constant k off
The freeze-thaw supernatant of the BCMA extracellular domain specific binding clone screened in the above step was analyzed using an Octet K2 molecular interaction analyzer. Biotinylated BCMA-huIgG1 Fc fusion protein was immobilized on SA probes and affinity assays were performed using freeze-thaw supernatants as analytes. In the analysis step, only the dissociation curve is selected to calculate the k off value, and the k off value is used as a reference basis to order from small to large. Preferably, clones k off, such as clones MC12, MB09, MB11, MA04, and MD07, order the top five.
Example 6: preparation of chimeric antigen receptor-T cells against human BCMA and determination of chimeric antigen receptor positive rate
1. Preparation of lentivirus packaging master plasmid containing CAR element
The antibody sequences of clones MB09, MC12, MD07 and HK10 and clone C11D5.3 were selected to construct chimeric antigen receptors. Construction of a CAR element lentiviral packaging master plasmid comprising the following structure from N-terminal to C-terminal by molecular cloning: a CD8 a signal peptide, an scFv antibody, a CD8 a hinge region, a CD8 a transmembrane region, a 41BB cytoplasmic region, and a CD3z cytoplasmic region. Clones sequenced correctly were selected, inoculated with bacterial solution into 300ml of 2YT medium, shaken overnight and the large plasmid was completed according to NucleoBond Xtra Maxi EF kit instructions.
2. Lentivirus package
Packaging the lentivirus-containing package with cationic polymer PEI, the procedure is as follows: PEI and lentiviral packaging plasmids (lentiviral master plasmid, RRE-SIV, REV, VSVG) were diluted with serum-free DMEM, respectively; adding PEI/DMEM into the plasmid/DMEM mixture, vortex shaking and mixing uniformly, and standing for 15 minutes at room temperature; the plasmid-PEI complex was added to pre-plated 293T cells (China academy of sciences cell bank). The liquid is changed 16h after transfection, virus supernatant is collected 48h later, filtered by a 0.45um filter, partial stock solution is reserved, and the rest is concentrated 30 times.
3. Virus titer assay
3.1 Biological titer determination
The biological titer of a virus solution refers to the number of infectious virus particles contained per milliliter.
Taking a 24-hole culture plate, adding a gradient diluted virus stock solution into each hole, wherein the initial infection volume is 1ml, the ratio of three times, five gradient dilutions, the volume is less than 1ml, and supplementing 1ml with a culture medium. After each well 100 μl of a cell suspension containing 1x10 5 293T cells (polybrene (Santa Cruz) at a final concentration of 8 μg/ml) was added, and 3 days later the efficiency of cell infection was measured by flow cytometry, the first staining using biotin-goat anti-mouse Fab (or biotin-goat anti-human IgG Fab), the second staining using PE-streptavidin to label the test cells, and uninfected, simultaneous culture of 293T cells as a negative control (293T). The titer calculation formula is: titer (TU/ml) =number of infected cells x flow assay CAR expression ratio/virus solution volume. The titer should be no less than 1X10 5 TU/ml.
The biological titer determination results were: the biological titer of the lentiviral stock solution containing the MB09 antibody is 24.6x10 6 TU/ml, the biological titer of the lentiviral stock solution containing the MC12 antibody is 3.06x10 6 TU/ml, the biological titer of the lentiviral stock solution containing the MD07 antibody is 2.10x10 6 TU/ml, the biological titer of the lentiviral stock solution containing the HK10 antibody is 8.82x10 6 TU/ml, and the biological titer of the lentiviral stock solution containing the C1 1D5.3 antibody is 2.65x10 6 TU/ml.
3.2 Physical titre determination
The physical titer of the virus solution is determined by the number of virus particles in the virus solution, including virus particles with infectious capability and defective virus particles without infectious capability. Lentiviral genetic material is known to be two single-stranded RNA genomes, and physical titer detection takes the CAR gene copy number in virus liquid as an evaluation index, and theoretically, two CAR gene copy numbers correspond to one virus particle.
Specifically, the present experiment uses the 41BB sequence of lentiviral gene as the amplification target to determine the physical titer of lentivirus. The virus supernatant was first DNA-treated and then subjected to one-step RT-PCR (TaKaRa) using 41BB specific fluorescent quantitative PCR primers to determine the copy number of the 41BB gene to determine the lentiviral genome copy number.
The physical titer test results were: the physical titer of the lentiviral stock solution containing the MB09 antibody is 4.65x10 6 copies/ul, the physical titer of the lentiviral stock solution containing the MC12 antibody is 5.99x10 6 copies/ul, the physical titer of the lentiviral stock solution containing the MD07 antibody is 6.89x10 6 copies/ul, the physical titer of the lentiviral stock solution containing the HK10 antibody is 3.21x10 6 copies/ul, and the physical titer of the lentiviral stock solution containing the C11D5.3 antibody is 4.26x10 6 copies/ul.
4. Preparation of chimeric antigen receptor-T cells and determination of chimeric antigen receptor-positive Rate
4.1CD3+T cell isolation and activation
Relatively pure CD3+ T cells were isolated using Ficcol% AB serum X-VIVO (LONZA) medium to a cell density of 1X10 6/mL. Cells were inoculated at 1 ml/well into cells previously infected with anti-human 50ng/ml CD3 antibody (Beijing co-dried sea cell) and 50ng/ml CD28 antibody (Beijing co-dried sea cell), followed by addition of 100IU/ml IL2 (Beijing Shuanglu), and after 48 hours of culture stimulation.
4.2 Infection with Virus stock and culture
The activated T cells were adjusted to 5X10 5/mL, 1mL of T cells and 1mL of virus stock were added to each well of a 24-well plate, and 1ul polybrene was added to each well, and centrifuged at 32℃and 2500rpm for 1.5 hours. The supernatant was discarded and 1ml of T cell medium (containing IL-250 IU/ml) was added to each well. The plates were placed in a 37℃5% CO 2 incubator for cultivation. 24h after infection, transferring to a 6-hole plate, observing the density of cells every day, and timely supplementing a T cell culture solution containing IL-250IU/ml to maintain the density of the T cells to be about 5x10 5/ml so as to expand the cells.
4.3CAR Positive Rate detection
CAR positive rate was detected 72h after virus infection. The chimeric antigen receptor group containing MB09, MC12, MD07, C11D5.3 clone and the negative control group are selected from biotin-goat anti-mouse IgG F (ab') 2 fragment as a primary antibody to incubate cells after lentivirus infection. The cells after lentiviral infection were incubated with biotin-goat anti-human IgG F (ab') 2 fragments as primary antibodies against the chimeric antigen receptor group containing the HK10 clone and the negative control group. After this time, the cells were incubated with Brilliant Violet 421. TM. Streptavidin (BioLegend) secondary antibody, and the 5 CAR-T positives were all substantially identical, at about 80%.
Example 7: functional verification of anti-human BCMA chimeric antigen receptor-T cells
1. Target cell construction
1.1 Packaging of lentiviruses
Luciferase and green fluorescent protein genes in tandem with Internal Ribosome Entry Sites (IRES) were synthesized by chemical synthesis. A lentivirus packaging master plasmid (PCCL-LUC-GFP) was constructed by molecular cloning and lentiviruses were packaged.
1.2K562-LUC-GFP and L363-LUC-GFP target cell construction
K562 (BCMA is not expressed, cell bank of China academy of sciences) and L363 (BCMA is expressed, cell bank of China academy of sciences) are respectively transfected by corresponding lentiviruses, and then the K562-LUC-GFP target cells and the L363-LUC-GFP target cells which are high in expression of luciferase and green fluorescent protein are screened out by taking green fluorescent protein as a marker.
2. Anti-human BCMA chimeric antigen receptor-T cell killing experiments
CAR-T killing experiments CAR-T cell in vitro function was assessed by detecting the killing effect of CAR-T cells on target cells in vitro. T cells were co-cultured with K562-LUC-GFP target cells and L363-LUC-GFP target cells, respectively, at different potency target ratios (3 x10 4 target cells with 1.5x10 5 effector cells and 3x10 4 target cells with 6x10 4 effector cells), while negative control groups of target cells mixed with untransfected CAR element T cells were set. After 18 hours, a luciferase reaction substrate was added to the culture system, and the fluorescence value was detected, and the killing efficiency was calculated by the following formula: killing efficiency = (1-experimental Kong Yingguang value/control Kong Yingguang value) x100%. The experimental groupings and results are shown in the following table.
In the cell killing experiments, 5 CAR-T and negative control T cells were essentially non-functional for K562. MB09, MC12, HK10 clones performed well for L363, with small differences in value compared to the positive control clone C11D5.3, but with weak MD07 clone. The results are shown in FIG. 1.
3. Anti-human BCMA chimeric antigen receptor-T cell IFN-gamma secretion and CD107a expression assay
After co-incubation of effector cells (CAR-T and negative control T cells without transfected CAR elements) with target cells (both effector and target cells are 3x10 5), their IFN- γ and CD107a expression were flow tested to evaluate CAR-T cell effector function in vitro after stimulation by target cells. The experimental groups are shown in the table, and after the effector cells and the target cells are mixed and placed in a 37 ℃ and co-incubated in a 5% CO 2 incubator for 4 hours, the proportion of cells secreting IFN-gamma and expressing CD107a in the CD3+ cells in each group of samples is detected, and the experimental groups and the results are shown in the table below.
Group of | CAR-T clone number | Target cells | CD3+IFN-γ+ | CD3+CD107a+ |
1 | MB09 | K562 | 1.30% | 0.91% |
2 | MC12 | K562 | 2.51% | 0.78% |
3 | MD07 | K562 | 7.97% | 6.83% |
4 | C11D5.3 | K562 | 2.45% | 0.82% |
5 | HK10 | K562 | 2.54% | 0.83% |
6 | NT | K562 | 1.33% | 0.68% |
7 | MB09 | L363 | 19.30% | 26.90% |
8 | MC12 | L363 | 18.90% | 25.00% |
9 | MD07 | L363 | 11.60% | 7.25% |
10 | C11D5.3 | L363 | 24.80% | 34.00% |
11 | HK10 | L363 | 34.40% | 44.60% |
12 | NT | L363 | 2.45% | 0.50% |
After co-incubation of CAR-T cells with target cells, MD07 clones stimulated non-specifically for K562 and L363, and both IFN- γ and CD107a expression were seen in CD3 positive cells. The remaining CAR-T and negative control T cells were non-functional for K562. HK10 was functionally stronger for L363 than the positive control clone C11D5.3, with MB09, MC12 being slightly weaker than the control. The results are shown in FIGS. 2-4.
Claims (10)
1. An antibody or antigen-binding fragment that specifically binds to human B Cell Maturation Antigen (BCMA), comprising a heavy chain variable domain and a light chain variable domain, said heavy chain variable domain and light chain variable domain comprising:
(1) SEQ ID NO:1, a heavy chain CDR1, SEQ ID NO:2, a heavy chain CDR2, SEQ ID NO:3, a heavy chain CDR3, SEQ ID NO:4, light chain CDR1, SEQ ID NO:5 and the light chain CDR2 shown in SEQ ID NO:6, a light chain CDR3;
(2) SEQ ID NO:7, heavy chain CDR1, SEQ ID NO:8, a heavy chain CDR2, SEQ ID NO:9, heavy chain CDR3, SEQ ID NO:10, light chain CDR1, SEQ ID NO:11 and the light chain CDR2 and SEQ ID NO:12, a light chain CDR3;
(3) SEQ ID NO:13, a heavy chain CDR1, SEQ ID NO:14, a heavy chain CDR2, SEQ ID NO:15, a heavy chain CDR3, SEQ ID NO:16, light chain CDR1, SEQ ID NO:17 and the light chain CDR2 and SEQ ID NO:18, a light chain CDR3;
(4) SEQ ID NO:19, a heavy chain CDR1, SEQ ID NO:20, a heavy chain CDR2, SEQ ID NO:21, heavy chain CDR3, SEQ ID NO:22, light chain CDR1, SEQ ID NO:23 and the light chain CDR2 shown in SEQ ID NO:24, a light chain CDR3; or (b)
(5) SEQ ID NO:25, a heavy chain CDR1, SEQ ID NO:26, a heavy chain CDR2, SEQ ID NO:27, heavy chain CDR3, SEQ ID NO:28, light chain CDR1, SEQ ID NO:29 and the light chain CDR2 and SEQ ID NO:30, and a light chain CDR3.
2. An antibody or antigen binding fragment that specifically binds to human B Cell Maturation Antigen (BCMA), comprising a heavy chain variable domain and a light chain variable domain comprising:
(1) SEQ ID NO:31, a heavy chain CDR1, SEQ ID NO:32, a heavy chain CDR2, SEQ ID NO:33, a heavy chain CDR3, SEQ ID NO:34, light chain CDR1, SEQ ID NO:35 and the light chain CDR2 and SEQ ID NO:36, a light chain CDR3;
(2) SEQ ID NO:37, a heavy chain CDR1, SEQ ID NO:38, the heavy chain CDR2, SEQ ID NO:39, heavy chain CDR3, SEQ ID NO:40, light chain CDR1, SEQ ID NO:41 and the light chain CDR2 and SEQ ID NO:42, a light chain CDR3;
(3) SEQ ID NO:43, a heavy chain CDR1, SEQ ID NO:44, a heavy chain CDR2, SEQ ID NO:45, heavy chain CDR3, SEQ ID NO:46, light chain CDR1, SEQ ID NO:47 and the light chain CDR2 and SEQ ID NO:48, a light chain CDR3;
(4) SEQ ID NO:49, heavy chain CDR1, SEQ ID NO:50, a heavy chain CDR2, SEQ ID NO:51, heavy chain CDR3, SEQ ID NO:52, light chain CDR1, SEQ ID NO:53 and the light chain CDR2 and SEQ ID NO:54 light chain CDR3; or (b)
(5) SEQ ID NO:55, heavy chain CDR1, SEQ ID NO:56, heavy chain CDR2, SEQ ID NO:57, a heavy chain CDR3, SEQ ID NO:58, light chain CDR1, SEQ ID NO:59 and light chain CDR2 and SEQ ID NO:60, and a light chain CDR3.
3. A chimeric antigen receptor comprising an extracellular region, a transmembrane region, and an intracellular region, wherein the extracellular region comprises an antigen binding region comprising a heavy chain variable domain and a light chain variable domain, the heavy chain variable domain and the light chain variable domain comprising:
(1) SEQ ID NO:1, a heavy chain CDR1, SEQ ID NO:2, a heavy chain CDR2, SEQ ID NO:3, a heavy chain CDR3, SEQ ID NO:4, light chain CDR1, SEQ ID NO:5 and the light chain CDR2 shown in SEQ ID NO:6, a light chain CDR3;
(2) SEQ ID NO:7, heavy chain CDR1, SEQ ID NO:8, a heavy chain CDR2, SEQ ID NO:9, heavy chain CDR3, SEQ ID NO:10, light chain CDR1, SEQ ID NO:11 and the light chain CDR2 and SEQ ID NO:12, a light chain CDR3;
(3) SEQ ID NO:13, a heavy chain CDR1, SEQ ID NO:14, a heavy chain CDR2, SEQ ID NO:15, a heavy chain CDR3, SEQ ID NO:16, light chain CDR1, SEQ ID NO:17 and the light chain CDR2 and SEQ ID NO:18, a light chain CDR3;
(4) SEQ ID NO:19, a heavy chain CDR1, SEQ ID NO:20, a heavy chain CDR2, SEQ ID NO:21, heavy chain CDR3, SEQ ID NO:22, light chain CDR1, SEQ ID NO:23 and the light chain CDR2 shown in SEQ ID NO:24, a light chain CDR3;
(5) SEQ ID NO:25, a heavy chain CDR1, SEQ ID NO:26, a heavy chain CDR2, SEQ ID NO:27, heavy chain CDR3, SEQ ID NO:28, light chain CDR1, SEQ ID NO:29 and the light chain CDR2 and SEQ ID NO:30, a light chain CDR3;
(6) SEQ ID NO:31, a heavy chain CDR1, SEQ ID NO:32, a heavy chain CDR2, SEQ ID NO:33, a heavy chain CDR3, SEQ ID NO:34, light chain CDR1, SEQ ID NO:35 and the light chain CDR2 and SEQ ID NO:36, a light chain CDR3;
(7) SEQ ID NO:37, a heavy chain CDR1, SEQ ID NO:38, the heavy chain CDR2, SEQ ID NO:39, heavy chain CDR3, SEQ ID NO:40, light chain CDR1, SEQ ID NO:41 and the light chain CDR2 and SEQ ID NO:42, a light chain CDR3;
(8) SEQ ID NO:43, a heavy chain CDR1, SEQ ID NO:44, a heavy chain CDR2, SEQ ID NO:45, heavy chain CDR3, SEQ ID NO:46, light chain CDR1, SEQ ID NO:47 and the light chain CDR2 and SEQ ID NO:48, a light chain CDR3;
(9) SEQ ID NO:49, heavy chain CDR1, SEQ ID NO:50, a heavy chain CDR2, SEQ ID NO:51, heavy chain CDR3, SEQ ID NO:52, light chain CDR1, SEQ ID NO:53 and the light chain CDR2 and SEQ ID NO:54 light chain CDR3; or (b)
(10) SEQ ID NO:55, heavy chain CDR1, SEQ ID NO:56, heavy chain CDR2, SEQ ID NO:57, a heavy chain CDR3, SEQ ID NO:58, light chain CDR1, SEQ ID NO:59 and light chain CDR2 and SEQ ID NO:60, and a light chain CDR3.
4. The chimeric antigen receptor according to claim 3, wherein the extracellular region further comprises a polypeptide having the amino acid sequence of SEQ ID NO:63, and a hinge region of the amino acid sequence shown in seq id no.
5. The chimeric antigen receptor according to claim 3 or 4, wherein the transmembrane region has the amino acid sequence of SEQ ID NO: 65.
6. The chimeric antigen receptor according to any one of claims 3 to 5, wherein the intracellular region has the sequence of SEQ ID NO:67 and/or the amino acid sequence shown in SEQ ID NO: 69.
7. A polynucleotide encoding an antibody or antigen binding fragment according to claim 1 or 2 or a chimeric antigen receptor according to any one of claims 3 to 6.
8. An engineered immune effector cell characterized in that it expresses the chimeric antigen receptor according to any one of claims 3 to 6, or comprises a polynucleotide encoding the chimeric antigen receptor according to any one of claims 3 to 6.
9. Use of an antibody or antigen binding fragment according to claim 1 or 2, a chimeric antigen receptor according to any one of claims 3 to 6, a polynucleotide according to claim 7, or an engineered immune effector cell according to claim 8 in the manufacture of a medicament for the treatment of cancer.
10. Use of an antibody or antigen binding fragment according to claim 1 or 2, a chimeric antigen receptor according to any one of claims 3 to 6, a polynucleotide according to claim 7, or an engineered immune effector cell according to claim 8 in the manufacture of a medicament for stimulating immune function in a patient with cancer.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202410294964.5A CN118206658A (en) | 2021-08-10 | 2021-08-10 | Chimeric antigen receptor of targeted BCMA based on fully human and murine single chain antibodies and application thereof |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202410294964.5A CN118206658A (en) | 2021-08-10 | 2021-08-10 | Chimeric antigen receptor of targeted BCMA based on fully human and murine single chain antibodies and application thereof |
CN202110912161.8A CN115703836A (en) | 2021-08-10 | 2021-08-10 | BCMA (brain cell activating antigen) targeted chimeric antigen receptor based on fully human and murine single-chain antibodies and application thereof |
Related Parent Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202110912161.8A Division CN115703836A (en) | 2021-08-10 | 2021-08-10 | BCMA (brain cell activating antigen) targeted chimeric antigen receptor based on fully human and murine single-chain antibodies and application thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN118206658A true CN118206658A (en) | 2024-06-18 |
Family
ID=85179463
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202410294964.5A Pending CN118206658A (en) | 2021-08-10 | 2021-08-10 | Chimeric antigen receptor of targeted BCMA based on fully human and murine single chain antibodies and application thereof |
CN202110912161.8A Pending CN115703836A (en) | 2021-08-10 | 2021-08-10 | BCMA (brain cell activating antigen) targeted chimeric antigen receptor based on fully human and murine single-chain antibodies and application thereof |
Family Applications After (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202110912161.8A Pending CN115703836A (en) | 2021-08-10 | 2021-08-10 | BCMA (brain cell activating antigen) targeted chimeric antigen receptor based on fully human and murine single-chain antibodies and application thereof |
Country Status (2)
Country | Link |
---|---|
CN (2) | CN118206658A (en) |
WO (1) | WO2023016576A1 (en) |
Family Cites Families (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN108004259B (en) * | 2016-11-02 | 2020-06-30 | 上海恒润达生生物科技有限公司 | Chimeric antigen receptor targeting B cell maturation antigen and uses thereof |
CN108395478A (en) * | 2017-02-04 | 2018-08-14 | 上海恒润达生生物科技有限公司 | Target the Chimeric antigen receptor of BCMA and the method and application thereof to its dual modification |
CN109320615B (en) * | 2018-09-25 | 2023-09-22 | 上海恒润达生生物科技股份有限公司 | Chimeric antigen receptor targeting novel BCMA and uses thereof |
US12116415B2 (en) * | 2018-10-09 | 2024-10-15 | Single Cell Technology, Inc. | Anti-BCMA antibodies |
CN109485733B (en) * | 2018-12-28 | 2020-04-24 | 广州百暨基因科技有限公司 | Fully human anti-BCMA chimeric antigen receptor and application thereof |
AU2020279224A1 (en) * | 2019-05-21 | 2021-12-16 | Novartis Ag | Trispecific binding molecules against BCMA and uses thereof |
CN112062851B (en) * | 2019-06-11 | 2022-09-09 | 南京驯鹿医疗技术有限公司 | Antibody targeting BCMA chimeric antigen receptor and application thereof |
CN114222758A (en) * | 2019-08-20 | 2022-03-22 | 武汉华大吉诺因生物科技有限公司 | anti-BCMA antibodies and their use in the CAR-T field |
-
2021
- 2021-08-10 CN CN202410294964.5A patent/CN118206658A/en active Pending
- 2021-08-10 CN CN202110912161.8A patent/CN115703836A/en active Pending
-
2022
- 2022-09-07 WO PCT/CN2022/117508 patent/WO2023016576A1/en active Application Filing
Also Published As
Publication number | Publication date |
---|---|
WO2023016576A1 (en) | 2023-02-16 |
CN115703836A (en) | 2023-02-17 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN108603200B (en) | Optimized lentiviral transfer vectors and uses thereof | |
JP2021525509A (en) | Diverse antigen-binding domains for cell therapy, new platforms and other enhancements | |
CN111629734A (en) | Novel platform for co-stimulation, novel CAR design and other enhancements of adoptive cell therapy | |
KR20190101979A (en) | Synthetic immune receptors and methods of use thereof | |
EP3964238A1 (en) | Bcma-targeting engineered immune cell and use thereof | |
CN113423726A (en) | Receptor providing targeted co-stimulation for adoptive cell therapy | |
US20220177524A1 (en) | Nef-containing t cells and methods of producing thereof | |
TW201930342A (en) | Multispecific chimeric receptors comprising an NKG2D domain and methods of use thereof | |
CN113039209A (en) | Compositions and methods for TCR reprogramming using fusion proteins | |
US20210324083A1 (en) | Methods and compositions comprising b7h3 chimeric antigen receptors | |
WO2021244654A1 (en) | Activation induced cytokine production in immune cells | |
US20230235067A1 (en) | SSTR-2 Binding Chimeric Antigen Receptors | |
CN116003628A (en) | Fully human specific chimeric antigen receptor targeting human DLL3 antigen and application thereof | |
US20220289814A1 (en) | Engineered t cells and methods of producing thereof | |
CN117157329A (en) | Fusion proteins and uses thereof | |
WO2021139755A1 (en) | Engineered t cell, and preparation and use thereof | |
WO2023072307A1 (en) | Antigen binding fragment targeting cd70, single-chain antibody and chimeric antigen receptor, and use thereof | |
CN109970859B (en) | Glyphican-3 specific antibody and CAR-T cell specific to the same | |
US20230226181A1 (en) | GENETIC ENGINEERING OF gamma delta T CELLS FOR IMMUNOTHERAPY | |
WO2023016576A1 (en) | Bcma-targeted chimeric antigen receptor based on fully human and mouse single-chain antibody and use thereof | |
CN111683971A (en) | Pharmaceutical recombinant receptor compositions and methods | |
EP4370213A1 (en) | Chimeric molecules providing targeted costimulation for adoptive cell therapy | |
US20230338422A1 (en) | Engineering gamma delta t cells with interleukin-36 for immunotherapy | |
WO2024094004A1 (en) | Fully human antibody targeting cd123 and use thereof | |
WO2024153124A1 (en) | Modified primary t cell and use thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |