WO2023104099A1 - 靶向bcma的p329g抗体及其与嵌合抗原受体细胞的组合和应用 - Google Patents
靶向bcma的p329g抗体及其与嵌合抗原受体细胞的组合和应用 Download PDFInfo
- Publication number
- WO2023104099A1 WO2023104099A1 PCT/CN2022/137265 CN2022137265W WO2023104099A1 WO 2023104099 A1 WO2023104099 A1 WO 2023104099A1 CN 2022137265 W CN2022137265 W CN 2022137265W WO 2023104099 A1 WO2023104099 A1 WO 2023104099A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- amino acid
- cells
- antibody
- seq
- cdr
- Prior art date
Links
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 title description 148
- 230000008685 targeting Effects 0.000 title description 9
- 101100425747 Mus musculus Tnfrsf17 gene Proteins 0.000 title description 5
- 210000003370 receptor cell Anatomy 0.000 title 1
- 210000004027 cell Anatomy 0.000 claims abstract description 594
- 108010008014 B-Cell Maturation Antigen Proteins 0.000 claims abstract description 287
- 102000006942 B-Cell Maturation Antigen Human genes 0.000 claims abstract description 285
- 210000001744 T-lymphocyte Anatomy 0.000 claims abstract description 99
- 108090000765 processed proteins & peptides Proteins 0.000 claims abstract description 73
- 229920001184 polypeptide Polymers 0.000 claims abstract description 68
- 102000004196 processed proteins & peptides Human genes 0.000 claims abstract description 68
- 150000007523 nucleic acids Chemical class 0.000 claims abstract description 66
- 239000013598 vector Substances 0.000 claims abstract description 65
- 108020004707 nucleic acids Proteins 0.000 claims abstract description 59
- 102000039446 nucleic acids Human genes 0.000 claims abstract description 59
- 238000011282 treatment Methods 0.000 claims abstract description 55
- 239000012642 immune effector Substances 0.000 claims abstract description 44
- 229940121354 immunomodulator Drugs 0.000 claims abstract description 44
- 230000001105 regulatory effect Effects 0.000 claims abstract description 44
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 38
- 230000035772 mutation Effects 0.000 claims abstract description 38
- 201000010099 disease Diseases 0.000 claims abstract description 35
- 210000000822 natural killer cell Anatomy 0.000 claims abstract description 34
- 102100035360 Cerebellar degeneration-related antigen 1 Human genes 0.000 claims description 171
- 101710117290 Aldo-keto reductase family 1 member C4 Proteins 0.000 claims description 170
- 150000001413 amino acids Chemical group 0.000 claims description 154
- 206010028980 Neoplasm Diseases 0.000 claims description 138
- 230000027455 binding Effects 0.000 claims description 134
- 241000282414 Homo sapiens Species 0.000 claims description 77
- 239000000427 antigen Substances 0.000 claims description 73
- 108091007433 antigens Proteins 0.000 claims description 73
- 102000036639 antigens Human genes 0.000 claims description 73
- 238000000034 method Methods 0.000 claims description 66
- 230000008859 change Effects 0.000 claims description 65
- GOJUJUVQIVIZAV-UHFFFAOYSA-N 2-amino-4,6-dichloropyrimidine-5-carbaldehyde Chemical group NC1=NC(Cl)=C(C=O)C(Cl)=N1 GOJUJUVQIVIZAV-UHFFFAOYSA-N 0.000 claims description 48
- 230000037396 body weight Effects 0.000 claims description 48
- 239000012634 fragment Substances 0.000 claims description 40
- 238000001727 in vivo Methods 0.000 claims description 33
- 230000011664 signaling Effects 0.000 claims description 33
- 238000006467 substitution reaction Methods 0.000 claims description 32
- NFGXHKASABOEEW-UHFFFAOYSA-N 1-methylethyl 11-methoxy-3,7,11-trimethyl-2,4-dodecadienoate Chemical compound COC(C)(C)CCCC(C)CC=CC(C)=CC(=O)OC(C)C NFGXHKASABOEEW-UHFFFAOYSA-N 0.000 claims description 30
- 206010035226 Plasma cell myeloma Diseases 0.000 claims description 30
- 230000004048 modification Effects 0.000 claims description 30
- 238000012986 modification Methods 0.000 claims description 30
- 230000001225 therapeutic effect Effects 0.000 claims description 30
- 208000034578 Multiple myelomas Diseases 0.000 claims description 28
- 201000011510 cancer Diseases 0.000 claims description 28
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 claims description 25
- 239000003814 drug Substances 0.000 claims description 20
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 claims description 19
- 108010047041 Complementarity Determining Regions Proteins 0.000 claims description 18
- 229940079593 drug Drugs 0.000 claims description 17
- 239000000890 drug combination Substances 0.000 claims description 16
- 230000000139 costimulatory effect Effects 0.000 claims description 14
- 238000012217 deletion Methods 0.000 claims description 13
- 230000037430 deletion Effects 0.000 claims description 13
- 230000004936 stimulating effect Effects 0.000 claims description 13
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 claims description 12
- 108010073807 IgG Receptors Proteins 0.000 claims description 12
- 108010076504 Protein Sorting Signals Proteins 0.000 claims description 12
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 claims description 12
- 238000007792 addition Methods 0.000 claims description 12
- 230000000735 allogeneic effect Effects 0.000 claims description 12
- 238000012544 monitoring process Methods 0.000 claims description 11
- 239000004471 Glycine Substances 0.000 claims description 10
- 102000009490 IgG Receptors Human genes 0.000 claims description 10
- 238000002360 preparation method Methods 0.000 claims description 10
- 230000001177 retroviral effect Effects 0.000 claims description 10
- 125000006850 spacer group Chemical group 0.000 claims description 9
- 230000002829 reductive effect Effects 0.000 claims description 8
- 239000013612 plasmid Substances 0.000 claims description 6
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 5
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 claims description 4
- 230000033228 biological regulation Effects 0.000 claims description 2
- 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 claims 13
- 108090000623 proteins and genes Proteins 0.000 abstract description 31
- 102000004169 proteins and genes Human genes 0.000 abstract description 14
- 235000001014 amino acid Nutrition 0.000 description 130
- 229940024606 amino acid Drugs 0.000 description 117
- 125000003275 alpha amino acid group Chemical group 0.000 description 95
- 241000699670 Mus sp. Species 0.000 description 93
- 210000004881 tumor cell Anatomy 0.000 description 58
- 230000000259 anti-tumor effect Effects 0.000 description 51
- 230000000694 effects Effects 0.000 description 49
- 241000699666 Mus <mouse, genus> Species 0.000 description 46
- 230000014509 gene expression Effects 0.000 description 40
- 238000001514 detection method Methods 0.000 description 38
- 230000002147 killing effect Effects 0.000 description 32
- 230000006870 function Effects 0.000 description 29
- 239000000306 component Substances 0.000 description 28
- 210000005259 peripheral blood Anatomy 0.000 description 27
- 239000011886 peripheral blood Substances 0.000 description 27
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 26
- 239000000872 buffer Substances 0.000 description 25
- 239000006285 cell suspension Substances 0.000 description 25
- 230000004913 activation Effects 0.000 description 24
- 239000012636 effector Substances 0.000 description 24
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 23
- 239000000203 mixture Substances 0.000 description 22
- 210000001519 tissue Anatomy 0.000 description 22
- 125000000539 amino acid group Chemical group 0.000 description 20
- 102000004127 Cytokines Human genes 0.000 description 19
- 108090000695 Cytokines Proteins 0.000 description 19
- 238000002474 experimental method Methods 0.000 description 19
- 239000000243 solution Substances 0.000 description 19
- 239000013604 expression vector Substances 0.000 description 17
- 241000713666 Lentivirus Species 0.000 description 16
- 230000005888 antibody-dependent cellular phagocytosis Effects 0.000 description 16
- 230000001965 increasing effect Effects 0.000 description 16
- 238000011081 inoculation Methods 0.000 description 16
- 241000894007 species Species 0.000 description 16
- 108091008874 T cell receptors Proteins 0.000 description 15
- 239000006228 supernatant Substances 0.000 description 15
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 14
- 210000004369 blood Anatomy 0.000 description 14
- 239000008280 blood Substances 0.000 description 14
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 13
- 238000000684 flow cytometry Methods 0.000 description 13
- 235000018102 proteins Nutrition 0.000 description 13
- 230000004044 response Effects 0.000 description 13
- 230000001404 mediated effect Effects 0.000 description 12
- 239000002609 medium Substances 0.000 description 12
- 230000035755 proliferation Effects 0.000 description 12
- 230000001988 toxicity Effects 0.000 description 12
- 231100000419 toxicity Toxicity 0.000 description 12
- 210000003462 vein Anatomy 0.000 description 12
- 230000001419 dependent effect Effects 0.000 description 11
- 238000000338 in vitro Methods 0.000 description 11
- 230000004068 intracellular signaling Effects 0.000 description 11
- 239000003446 ligand Substances 0.000 description 11
- 230000000638 stimulation Effects 0.000 description 11
- 108060003951 Immunoglobulin Proteins 0.000 description 10
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 10
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 10
- 102000018358 immunoglobulin Human genes 0.000 description 10
- 239000007924 injection Substances 0.000 description 10
- 238000002347 injection Methods 0.000 description 10
- 239000000523 sample Substances 0.000 description 10
- 210000002966 serum Anatomy 0.000 description 10
- 238000003556 assay Methods 0.000 description 9
- 230000004540 complement-dependent cytotoxicity Effects 0.000 description 9
- 108020001507 fusion proteins Proteins 0.000 description 9
- 102000037865 fusion proteins Human genes 0.000 description 9
- 238000001802 infusion Methods 0.000 description 9
- 230000003834 intracellular effect Effects 0.000 description 9
- 241000282693 Cercopithecidae Species 0.000 description 8
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 8
- 108091028043 Nucleic acid sequence Proteins 0.000 description 8
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 8
- 210000003719 b-lymphocyte Anatomy 0.000 description 8
- 239000011324 bead Substances 0.000 description 8
- 231100000331 toxic Toxicity 0.000 description 8
- 230000002588 toxic effect Effects 0.000 description 8
- 238000005406 washing Methods 0.000 description 8
- 101150078244 AMO1 gene Proteins 0.000 description 7
- 241000283707 Capra Species 0.000 description 7
- 101000801255 Homo sapiens Tumor necrosis factor receptor superfamily member 17 Proteins 0.000 description 7
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 7
- 230000001086 cytosolic effect Effects 0.000 description 7
- 238000010790 dilution Methods 0.000 description 7
- 239000012895 dilution Substances 0.000 description 7
- 208000015181 infectious disease Diseases 0.000 description 7
- 230000007774 longterm Effects 0.000 description 7
- 239000000126 substance Substances 0.000 description 7
- 238000002965 ELISA Methods 0.000 description 6
- 108010002350 Interleukin-2 Proteins 0.000 description 6
- 102000000588 Interleukin-2 Human genes 0.000 description 6
- 241000282567 Macaca fascicularis Species 0.000 description 6
- 241001465754 Metazoa Species 0.000 description 6
- 241000283973 Oryctolagus cuniculus Species 0.000 description 6
- 101710138747 Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 6
- 241000283984 Rodentia Species 0.000 description 6
- 238000002617 apheresis Methods 0.000 description 6
- 230000008901 benefit Effects 0.000 description 6
- 238000010586 diagram Methods 0.000 description 6
- 238000010494 dissociation reaction Methods 0.000 description 6
- 230000005593 dissociations Effects 0.000 description 6
- 230000001939 inductive effect Effects 0.000 description 6
- 239000007928 intraperitoneal injection Substances 0.000 description 6
- 210000004962 mammalian cell Anatomy 0.000 description 6
- 210000004180 plasmocyte Anatomy 0.000 description 6
- 239000013641 positive control Substances 0.000 description 6
- 230000008569 process Effects 0.000 description 6
- 208000031648 Body Weight Changes Diseases 0.000 description 5
- 238000011357 CAR T-cell therapy Methods 0.000 description 5
- 108020004414 DNA Proteins 0.000 description 5
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 5
- 102000009109 Fc receptors Human genes 0.000 description 5
- 108010087819 Fc receptors Proteins 0.000 description 5
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 5
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 5
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 5
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 5
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 5
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 5
- 241000124008 Mammalia Species 0.000 description 5
- 206010029350 Neurotoxicity Diseases 0.000 description 5
- 241000700159 Rattus Species 0.000 description 5
- 206010044221 Toxic encephalopathy Diseases 0.000 description 5
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 5
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 230000004579 body weight change Effects 0.000 description 5
- 210000001185 bone marrow Anatomy 0.000 description 5
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 5
- 239000012141 concentrate Substances 0.000 description 5
- 206010052015 cytokine release syndrome Diseases 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 231100000673 dose–response relationship Toxicity 0.000 description 5
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 5
- 102000046935 human TNFRSF17 Human genes 0.000 description 5
- 230000007135 neurotoxicity Effects 0.000 description 5
- 231100000228 neurotoxicity Toxicity 0.000 description 5
- 102000005962 receptors Human genes 0.000 description 5
- 108020003175 receptors Proteins 0.000 description 5
- 230000028327 secretion Effects 0.000 description 5
- 230000019491 signal transduction Effects 0.000 description 5
- 230000004083 survival effect Effects 0.000 description 5
- 238000003786 synthesis reaction Methods 0.000 description 5
- 238000012546 transfer Methods 0.000 description 5
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 5
- 239000004474 valine Substances 0.000 description 5
- 235000014393 valine Nutrition 0.000 description 5
- FYELSNVLZVIGTI-UHFFFAOYSA-N 2-[4-[2-(2,3-dihydro-1H-inden-2-ylamino)pyrimidin-5-yl]-5-ethylpyrazol-1-yl]-1-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)ethanone Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C=1C=NN(C=1CC)CC(=O)N1CC2=C(CC1)NN=N2 FYELSNVLZVIGTI-UHFFFAOYSA-N 0.000 description 4
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 4
- 108091007741 Chimeric antigen receptor T cells Proteins 0.000 description 4
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 4
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 4
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 4
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 4
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 4
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 4
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 4
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 4
- 230000006044 T cell activation Effects 0.000 description 4
- 210000000662 T-lymphocyte subset Anatomy 0.000 description 4
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 4
- 239000004473 Threonine Substances 0.000 description 4
- 108700019146 Transgenes Proteins 0.000 description 4
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- 101150063416 add gene Proteins 0.000 description 4
- 230000003698 anagen phase Effects 0.000 description 4
- 230000004071 biological effect Effects 0.000 description 4
- 239000006143 cell culture medium Substances 0.000 description 4
- 230000010261 cell growth Effects 0.000 description 4
- 230000006037 cell lysis Effects 0.000 description 4
- 239000011248 coating agent Substances 0.000 description 4
- 238000000576 coating method Methods 0.000 description 4
- 230000024203 complement activation Effects 0.000 description 4
- 150000001875 compounds Chemical class 0.000 description 4
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Chemical compound CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 description 4
- 238000012258 culturing Methods 0.000 description 4
- 230000001472 cytotoxic effect Effects 0.000 description 4
- 239000012091 fetal bovine serum Substances 0.000 description 4
- 230000004927 fusion Effects 0.000 description 4
- 230000036541 health Effects 0.000 description 4
- 235000014705 isoleucine Nutrition 0.000 description 4
- 229960000310 isoleucine Drugs 0.000 description 4
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 4
- 210000003292 kidney cell Anatomy 0.000 description 4
- 210000004698 lymphocyte Anatomy 0.000 description 4
- 238000004519 manufacturing process Methods 0.000 description 4
- 239000011159 matrix material Substances 0.000 description 4
- 210000004379 membrane Anatomy 0.000 description 4
- 239000012528 membrane Substances 0.000 description 4
- 239000002773 nucleotide Substances 0.000 description 4
- 125000003729 nucleotide group Chemical group 0.000 description 4
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 4
- 235000008729 phenylalanine Nutrition 0.000 description 4
- 229960005190 phenylalanine Drugs 0.000 description 4
- 239000000047 product Substances 0.000 description 4
- 235000013930 proline Nutrition 0.000 description 4
- 230000009467 reduction Effects 0.000 description 4
- 238000011160 research Methods 0.000 description 4
- 208000024891 symptom Diseases 0.000 description 4
- 238000001890 transfection Methods 0.000 description 4
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 4
- 230000003612 virological effect Effects 0.000 description 4
- 210000005253 yeast cell Anatomy 0.000 description 4
- VZSRBBMJRBPUNF-UHFFFAOYSA-N 2-(2,3-dihydro-1H-inden-2-ylamino)-N-[3-oxo-3-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)propyl]pyrimidine-5-carboxamide Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C(=O)NCCC(N1CC2=C(CC1)NN=N2)=O VZSRBBMJRBPUNF-UHFFFAOYSA-N 0.000 description 3
- JVKRKMWZYMKVTQ-UHFFFAOYSA-N 2-[4-[2-(2,3-dihydro-1H-inden-2-ylamino)pyrimidin-5-yl]pyrazol-1-yl]-N-(2-oxo-3H-1,3-benzoxazol-6-yl)acetamide Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C=1C=NN(C=1)CC(=O)NC1=CC2=C(NC(O2)=O)C=C1 JVKRKMWZYMKVTQ-UHFFFAOYSA-N 0.000 description 3
- IVLXQGJVBGMLRR-UHFFFAOYSA-N 2-aminoacetic acid;hydron;chloride Chemical compound Cl.NCC(O)=O IVLXQGJVBGMLRR-UHFFFAOYSA-N 0.000 description 3
- 229940124661 Abecma Drugs 0.000 description 3
- 206010003445 Ascites Diseases 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 206010061818 Disease progression Diseases 0.000 description 3
- 101100118093 Drosophila melanogaster eEF1alpha2 gene Proteins 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 108091006020 Fc-tagged proteins Proteins 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 3
- 101001023379 Homo sapiens Lysosome-associated membrane glycoprotein 1 Proteins 0.000 description 3
- 241000725303 Human immunodeficiency virus Species 0.000 description 3
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 3
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 3
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 3
- 102100029204 Low affinity immunoglobulin gamma Fc region receptor II-a Human genes 0.000 description 3
- 239000005089 Luciferase Substances 0.000 description 3
- 102100035133 Lysosome-associated membrane glycoprotein 1 Human genes 0.000 description 3
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 3
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 3
- 102100036922 Tumor necrosis factor ligand superfamily member 13B Human genes 0.000 description 3
- 230000003213 activating effect Effects 0.000 description 3
- 235000004279 alanine Nutrition 0.000 description 3
- 238000001574 biopsy Methods 0.000 description 3
- 230000000903 blocking effect Effects 0.000 description 3
- 101150058049 car gene Proteins 0.000 description 3
- 230000020411 cell activation Effects 0.000 description 3
- 210000000170 cell membrane Anatomy 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 238000004587 chromatography analysis Methods 0.000 description 3
- 230000004154 complement system Effects 0.000 description 3
- 230000008878 coupling Effects 0.000 description 3
- 238000010168 coupling process Methods 0.000 description 3
- 238000005859 coupling reaction Methods 0.000 description 3
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 3
- 235000018417 cysteine Nutrition 0.000 description 3
- 230000009089 cytolysis Effects 0.000 description 3
- 231100000433 cytotoxic Toxicity 0.000 description 3
- 230000003013 cytotoxicity Effects 0.000 description 3
- 231100000135 cytotoxicity Toxicity 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 230000005750 disease progression Effects 0.000 description 3
- 208000035475 disorder Diseases 0.000 description 3
- 230000003828 downregulation Effects 0.000 description 3
- 239000013613 expression plasmid Substances 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 238000001476 gene delivery Methods 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 230000004727 humoral immunity Effects 0.000 description 3
- 108700004894 idecabtagene vicleucel Proteins 0.000 description 3
- 230000028993 immune response Effects 0.000 description 3
- 238000011534 incubation Methods 0.000 description 3
- 235000005772 leucine Nutrition 0.000 description 3
- 210000000265 leukocyte Anatomy 0.000 description 3
- 230000000670 limiting effect Effects 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 231100000682 maximum tolerated dose Toxicity 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 108091033319 polynucleotide Proteins 0.000 description 3
- 102000040430 polynucleotide Human genes 0.000 description 3
- 239000002157 polynucleotide Substances 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 208000037821 progressive disease Diseases 0.000 description 3
- 230000008929 regeneration Effects 0.000 description 3
- 238000011069 regeneration method Methods 0.000 description 3
- 238000012552 review Methods 0.000 description 3
- 238000013207 serial dilution Methods 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 230000004614 tumor growth Effects 0.000 description 3
- 241001430294 unidentified retrovirus Species 0.000 description 3
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 2
- PGOHTUIFYSHAQG-LJSDBVFPSA-N (2S)-6-amino-2-[[(2S)-5-amino-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-4-amino-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-5-amino-2-[[(2S)-5-amino-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S,3R)-2-[[(2S)-5-amino-2-[[(2S)-2-[[(2S)-2-[[(2S,3R)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-5-amino-2-[[(2S)-1-[(2S,3R)-2-[[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2S)-2-[[(2S)-2-[[2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-1-[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-amino-4-methylsulfanylbutanoyl]amino]-3-(1H-indol-3-yl)propanoyl]amino]-5-carbamimidamidopentanoyl]amino]propanoyl]pyrrolidine-2-carbonyl]amino]-3-methylbutanoyl]amino]-4-methylpentanoyl]amino]-4-methylpentanoyl]amino]acetyl]amino]-3-hydroxypropanoyl]amino]-4-methylpentanoyl]amino]-3-sulfanylpropanoyl]amino]-4-methylsulfanylbutanoyl]amino]-5-carbamimidamidopentanoyl]amino]-3-hydroxybutanoyl]pyrrolidine-2-carbonyl]amino]-5-oxopentanoyl]amino]-3-hydroxypropanoyl]amino]-3-hydroxypropanoyl]amino]-3-(1H-imidazol-5-yl)propanoyl]amino]-4-methylpentanoyl]amino]-3-hydroxybutanoyl]amino]-3-(1H-indol-3-yl)propanoyl]amino]-5-carbamimidamidopentanoyl]amino]-5-oxopentanoyl]amino]-3-hydroxybutanoyl]amino]-3-hydroxypropanoyl]amino]-3-carboxypropanoyl]amino]-3-hydroxypropanoyl]amino]-5-oxopentanoyl]amino]-5-oxopentanoyl]amino]-3-phenylpropanoyl]amino]-5-carbamimidamidopentanoyl]amino]-3-methylbutanoyl]amino]-4-methylpentanoyl]amino]-4-oxobutanoyl]amino]-5-carbamimidamidopentanoyl]amino]-3-(1H-indol-3-yl)propanoyl]amino]-4-carboxybutanoyl]amino]-5-oxopentanoyl]amino]hexanoic acid Chemical compound CSCC[C@H](N)C(=O)N[C@@H](Cc1c[nH]c2ccccc12)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N1CCC[C@H]1C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)NCC(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1CCC[C@H]1C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CO)C(=O)N[C@@H](Cc1cnc[nH]1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](Cc1c[nH]c2ccccc12)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](Cc1ccccc1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](Cc1c[nH]c2ccccc12)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCCN)C(O)=O PGOHTUIFYSHAQG-LJSDBVFPSA-N 0.000 description 2
- ZRPAUEVGEGEPFQ-UHFFFAOYSA-N 2-[4-[2-(2,3-dihydro-1H-inden-2-ylamino)pyrimidin-5-yl]pyrazol-1-yl]-1-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)ethanone Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C=1C=NN(C=1)CC(=O)N1CC2=C(CC1)NN=N2 ZRPAUEVGEGEPFQ-UHFFFAOYSA-N 0.000 description 2
- 102000006306 Antigen Receptors Human genes 0.000 description 2
- 108010083359 Antigen Receptors Proteins 0.000 description 2
- 239000004475 Arginine Substances 0.000 description 2
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 2
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 2
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 2
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 description 2
- 241000701022 Cytomegalovirus Species 0.000 description 2
- 108010021468 Fc gamma receptor IIA Proteins 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
- BCCRXDTUTZHDEU-VKHMYHEASA-N Gly-Ser Chemical compound NCC(=O)N[C@@H](CO)C(O)=O BCCRXDTUTZHDEU-VKHMYHEASA-N 0.000 description 2
- 102000001554 Hemoglobins Human genes 0.000 description 2
- 108010054147 Hemoglobins Proteins 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 2
- 101000581981 Homo sapiens Neural cell adhesion molecule 1 Proteins 0.000 description 2
- -1 IL-1α Proteins 0.000 description 2
- 206010061598 Immunodeficiency Diseases 0.000 description 2
- 108010074328 Interferon-gamma Proteins 0.000 description 2
- 102000015696 Interleukins Human genes 0.000 description 2
- 108010063738 Interleukins Proteins 0.000 description 2
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 2
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 2
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 2
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 2
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 2
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 2
- 102100029193 Low affinity immunoglobulin gamma Fc region receptor III-A Human genes 0.000 description 2
- 101710099301 Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 2
- 239000004472 Lysine Substances 0.000 description 2
- 101710085938 Matrix protein Proteins 0.000 description 2
- 101710127721 Membrane protein Proteins 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 2
- 102100027347 Neural cell adhesion molecule 1 Human genes 0.000 description 2
- 108010077524 Peptide Elongation Factor 1 Proteins 0.000 description 2
- 208000002151 Pleural effusion Diseases 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 2
- 241000705082 Sialia Species 0.000 description 2
- 108010000499 Thromboplastin Proteins 0.000 description 2
- 102100030859 Tissue factor Human genes 0.000 description 2
- 102100040247 Tumor necrosis factor Human genes 0.000 description 2
- 102100033726 Tumor necrosis factor receptor superfamily member 17 Human genes 0.000 description 2
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 2
- 239000002250 absorbent Substances 0.000 description 2
- 230000002745 absorbent Effects 0.000 description 2
- 230000002378 acidificating effect Effects 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 210000004381 amniotic fluid Anatomy 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 238000009175 antibody therapy Methods 0.000 description 2
- 230000000890 antigenic effect Effects 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 2
- 210000001106 artificial yeast chromosome Anatomy 0.000 description 2
- 125000003118 aryl group Chemical group 0.000 description 2
- 235000009582 asparagine Nutrition 0.000 description 2
- 229960001230 asparagine Drugs 0.000 description 2
- 235000003704 aspartic acid Nutrition 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 102000015736 beta 2-Microglobulin Human genes 0.000 description 2
- 108010081355 beta 2-Microglobulin Proteins 0.000 description 2
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 2
- 238000010876 biochemical test Methods 0.000 description 2
- 229960000074 biopharmaceutical Drugs 0.000 description 2
- 239000012503 blood component Substances 0.000 description 2
- 210000001772 blood platelet Anatomy 0.000 description 2
- 210000001124 body fluid Anatomy 0.000 description 2
- 239000010839 body fluid Substances 0.000 description 2
- 210000004899 c-terminal region Anatomy 0.000 description 2
- 238000004364 calculation method Methods 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 230000036755 cellular response Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 230000002759 chromosomal effect Effects 0.000 description 2
- 238000011260 co-administration Methods 0.000 description 2
- 238000003501 co-culture Methods 0.000 description 2
- 238000002648 combination therapy Methods 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 238000002591 computed tomography Methods 0.000 description 2
- 238000010276 construction Methods 0.000 description 2
- 238000007796 conventional method Methods 0.000 description 2
- 229940109239 creatinine Drugs 0.000 description 2
- 238000002784 cytotoxicity assay Methods 0.000 description 2
- 231100000263 cytotoxicity test Toxicity 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 238000003745 diagnosis Methods 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 210000003743 erythrocyte Anatomy 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 210000003722 extracellular fluid Anatomy 0.000 description 2
- 238000002073 fluorescence micrograph Methods 0.000 description 2
- 238000001502 gel electrophoresis Methods 0.000 description 2
- 235000013922 glutamic acid Nutrition 0.000 description 2
- 239000004220 glutamic acid Substances 0.000 description 2
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- 238000004128 high performance liquid chromatography Methods 0.000 description 2
- 230000005847 immunogenicity Effects 0.000 description 2
- 229940072221 immunoglobulins Drugs 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 238000007689 inspection Methods 0.000 description 2
- 230000010354 integration Effects 0.000 description 2
- 229940047122 interleukins Drugs 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 238000010253 intravenous injection Methods 0.000 description 2
- 238000004255 ion exchange chromatography Methods 0.000 description 2
- 238000001155 isoelectric focusing Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 230000003902 lesion Effects 0.000 description 2
- 210000005229 liver cell Anatomy 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 229930182817 methionine Natural products 0.000 description 2
- 235000006109 methionine Nutrition 0.000 description 2
- 244000005700 microbiome Species 0.000 description 2
- 230000009149 molecular binding Effects 0.000 description 2
- 201000000050 myeloid neoplasm Diseases 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 210000002381 plasma Anatomy 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 238000004393 prognosis Methods 0.000 description 2
- 210000001938 protoplast Anatomy 0.000 description 2
- 238000009256 replacement therapy Methods 0.000 description 2
- 238000002741 site-directed mutagenesis Methods 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000002459 sustained effect Effects 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 230000009261 transgenic effect Effects 0.000 description 2
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 2
- 210000002700 urine Anatomy 0.000 description 2
- 230000002861 ventricular Effects 0.000 description 2
- QZNNVYOVQUKYSC-JEDNCBNOSA-N (2s)-2-amino-3-(1h-imidazol-5-yl)propanoic acid;hydron;chloride Chemical compound Cl.OC(=O)[C@@H](N)CC1=CN=CN1 QZNNVYOVQUKYSC-JEDNCBNOSA-N 0.000 description 1
- 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 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- UKAUYVFTDYCKQA-UHFFFAOYSA-N -2-Amino-4-hydroxybutanoic acid Natural products OC(=O)C(N)CCO UKAUYVFTDYCKQA-UHFFFAOYSA-N 0.000 description 1
- WWSJZGAPAVMETJ-UHFFFAOYSA-N 2-[4-[2-(2,3-dihydro-1H-inden-2-ylamino)pyrimidin-5-yl]-3-ethoxypyrazol-1-yl]-1-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)ethanone Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C=1C(=NN(C=1)CC(=O)N1CC2=C(CC1)NN=N2)OCC WWSJZGAPAVMETJ-UHFFFAOYSA-N 0.000 description 1
- UAIUNKRWKOVEES-UHFFFAOYSA-N 3,3',5,5'-tetramethylbenzidine Chemical compound CC1=C(N)C(C)=CC(C=2C=C(C)C(N)=C(C)C=2)=C1 UAIUNKRWKOVEES-UHFFFAOYSA-N 0.000 description 1
- BRMWTNUJHUMWMS-UHFFFAOYSA-N 3-Methylhistidine Natural products CN1C=NC(CC(N)C(O)=O)=C1 BRMWTNUJHUMWMS-UHFFFAOYSA-N 0.000 description 1
- 229940117976 5-hydroxylysine Drugs 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 1
- 102000002659 Amyloid Precursor Protein Secretases Human genes 0.000 description 1
- 108010043324 Amyloid Precursor Protein Secretases Proteins 0.000 description 1
- 241000714230 Avian leukemia virus Species 0.000 description 1
- 108091008875 B cell receptors Proteins 0.000 description 1
- 108010028006 B-Cell Activating Factor Proteins 0.000 description 1
- 230000003844 B-cell-activation Effects 0.000 description 1
- 238000011725 BALB/c mouse Methods 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 102100027207 CD27 antigen Human genes 0.000 description 1
- 101150013553 CD40 gene Proteins 0.000 description 1
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 1
- 102100026550 Caspase-9 Human genes 0.000 description 1
- 108090000566 Caspase-9 Proteins 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 241000282552 Chlorocebus aethiops Species 0.000 description 1
- 208000005443 Circulating Neoplastic Cells Diseases 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 102000014447 Complement C1q Human genes 0.000 description 1
- 108010078043 Complement C1q Proteins 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- 102000004420 Creatine Kinase Human genes 0.000 description 1
- 108010042126 Creatine kinase Proteins 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 1
- RWSOTUBLDIXVET-UHFFFAOYSA-N Dihydrogen sulfide Chemical compound S RWSOTUBLDIXVET-UHFFFAOYSA-N 0.000 description 1
- 206010058314 Dysplasia Diseases 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000701959 Escherichia virus Lambda Species 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 108010021472 Fc gamma receptor IIB Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 108010049003 Fibrinogen Proteins 0.000 description 1
- 102000008946 Fibrinogen Human genes 0.000 description 1
- 229920001917 Ficoll Polymers 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 102100026122 High affinity immunoglobulin gamma Fc receptor I Human genes 0.000 description 1
- 102000008949 Histocompatibility Antigens Class I Human genes 0.000 description 1
- 241001272567 Hominoidea Species 0.000 description 1
- 101000690301 Homo sapiens Aldo-keto reductase family 1 member C4 Proteins 0.000 description 1
- 101100005238 Homo sapiens CARTPT gene Proteins 0.000 description 1
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 1
- 101000935587 Homo sapiens Flavin reductase (NADPH) Proteins 0.000 description 1
- 101000913074 Homo sapiens High affinity immunoglobulin gamma Fc receptor I Proteins 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- 101000917826 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-a Proteins 0.000 description 1
- 101000917824 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-b Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101001116548 Homo sapiens Protein CBFA2T1 Proteins 0.000 description 1
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 1
- 101000801234 Homo sapiens Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- PMMYEEVYMWASQN-DMTCNVIQSA-N Hydroxyproline Chemical compound O[C@H]1CN[C@H](C(O)=O)C1 PMMYEEVYMWASQN-DMTCNVIQSA-N 0.000 description 1
- 208000029462 Immunodeficiency disease Diseases 0.000 description 1
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 1
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 1
- 238000012404 In vitro experiment Methods 0.000 description 1
- 102100037850 Interferon gamma Human genes 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 108090000177 Interleukin-11 Proteins 0.000 description 1
- 108010065805 Interleukin-12 Proteins 0.000 description 1
- 108090000172 Interleukin-15 Proteins 0.000 description 1
- 108010002386 Interleukin-3 Proteins 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 108010002616 Interleukin-5 Proteins 0.000 description 1
- 108090001005 Interleukin-6 Proteins 0.000 description 1
- 108090001007 Interleukin-8 Proteins 0.000 description 1
- 108010002335 Interleukin-9 Proteins 0.000 description 1
- AHLPHDHHMVZTML-BYPYZUCNSA-N L-Ornithine Chemical compound NCCC[C@H](N)C(O)=O AHLPHDHHMVZTML-BYPYZUCNSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- UKAUYVFTDYCKQA-VKHMYHEASA-N L-homoserine Chemical compound OC(=O)[C@@H](N)CCO UKAUYVFTDYCKQA-VKHMYHEASA-N 0.000 description 1
- 102100029205 Low affinity immunoglobulin gamma Fc region receptor II-b Human genes 0.000 description 1
- 102000008072 Lymphokines Human genes 0.000 description 1
- 108010074338 Lymphokines Proteins 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 102000004083 Lymphotoxin-alpha Human genes 0.000 description 1
- 108090000542 Lymphotoxin-alpha Proteins 0.000 description 1
- 108091054437 MHC class I family Proteins 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 108010050619 Monokines Proteins 0.000 description 1
- 102000013967 Monokines Human genes 0.000 description 1
- 241000713333 Mouse mammary tumor virus Species 0.000 description 1
- 241000714177 Murine leukemia virus Species 0.000 description 1
- 101100066433 Mus musculus Fcgr3 gene Proteins 0.000 description 1
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- 102100032965 Myomesin-2 Human genes 0.000 description 1
- 102000003505 Myosin Human genes 0.000 description 1
- 108060008487 Myosin Proteins 0.000 description 1
- JDHILDINMRGULE-LURJTMIESA-N N(pros)-methyl-L-histidine Chemical compound CN1C=NC=C1C[C@H](N)C(O)=O JDHILDINMRGULE-LURJTMIESA-N 0.000 description 1
- 108010057466 NF-kappa B Proteins 0.000 description 1
- 102000003945 NF-kappa B Human genes 0.000 description 1
- 108091008877 NK cell receptors Proteins 0.000 description 1
- 108091007491 NSP3 Papain-like protease domains Proteins 0.000 description 1
- 102000010648 Natural Killer Cell Receptors Human genes 0.000 description 1
- 108010038807 Oligopeptides Proteins 0.000 description 1
- 102000015636 Oligopeptides Human genes 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- AHLPHDHHMVZTML-UHFFFAOYSA-N Orn-delta-NH2 Natural products NCCCC(N)C(O)=O AHLPHDHHMVZTML-UHFFFAOYSA-N 0.000 description 1
- UTJLXEIPEHZYQJ-UHFFFAOYSA-N Ornithine Natural products OC(=O)C(C)CCCN UTJLXEIPEHZYQJ-UHFFFAOYSA-N 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 102000010292 Peptide Elongation Factor 1 Human genes 0.000 description 1
- 229920002594 Polyethylene Glycol 8000 Polymers 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 206010036790 Productive cough Diseases 0.000 description 1
- 229940079156 Proteasome inhibitor Drugs 0.000 description 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 108010094028 Prothrombin Proteins 0.000 description 1
- 102100027378 Prothrombin Human genes 0.000 description 1
- 241000700157 Rattus norvegicus Species 0.000 description 1
- 241000712907 Retroviridae Species 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 102000008115 Signaling Lymphocytic Activation Molecule Family Member 1 Human genes 0.000 description 1
- 108010074687 Signaling Lymphocytic Activation Molecule Family Member 1 Proteins 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 102000006601 Thymidine Kinase Human genes 0.000 description 1
- 108020004440 Thymidine kinase Proteins 0.000 description 1
- 238000008050 Total Bilirubin Reagent Methods 0.000 description 1
- 102100037116 Transcription elongation factor 1 homolog Human genes 0.000 description 1
- 108020004566 Transfer RNA Proteins 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 1
- 102100033728 Tumor necrosis factor receptor superfamily member 18 Human genes 0.000 description 1
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 1
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 1
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 1
- 108091005966 Type III transmembrane proteins Proteins 0.000 description 1
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 108091093126 WHP Posttrascriptional Response Element Proteins 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 125000000266 alpha-aminoacyl group Chemical group 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 230000006229 amino acid addition Effects 0.000 description 1
- 150000003862 amino acid derivatives Chemical class 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000002924 anti-infective effect Effects 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 230000001446 anti-myeloma Effects 0.000 description 1
- 239000003146 anticoagulant agent Substances 0.000 description 1
- 229940127219 anticoagulant drug Drugs 0.000 description 1
- 238000000149 argon plasma sintering Methods 0.000 description 1
- 238000010420 art technique Methods 0.000 description 1
- 210000003567 ascitic fluid Anatomy 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 238000001815 biotherapy Methods 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 230000005907 cancer growth Effects 0.000 description 1
- 238000005251 capillar electrophoresis Methods 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000005859 cell recognition Effects 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 238000003570 cell viability assay Methods 0.000 description 1
- 238000012054 celltiter-glo Methods 0.000 description 1
- 230000010001 cellular homeostasis Effects 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 230000007541 cellular toxicity Effects 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 229940054315 ciltacabtagene autoleucel Drugs 0.000 description 1
- 230000009194 climbing Effects 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 238000005345 coagulation Methods 0.000 description 1
- 230000015271 coagulation Effects 0.000 description 1
- 108700032673 cocaine- and amphetamine-regulated transcript Proteins 0.000 description 1
- 238000011970 concomitant therapy Methods 0.000 description 1
- 239000003433 contraceptive agent Substances 0.000 description 1
- 230000002254 contraceptive effect Effects 0.000 description 1
- 229910052802 copper Inorganic materials 0.000 description 1
- 239000010949 copper Substances 0.000 description 1
- 239000002577 cryoprotective agent Substances 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- 102000003675 cytokine receptors Human genes 0.000 description 1
- 108010057085 cytokine receptors Proteins 0.000 description 1
- 230000001461 cytolytic effect Effects 0.000 description 1
- 230000003436 cytoskeletal effect Effects 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- YSMODUONRAFBET-UHFFFAOYSA-N delta-DL-hydroxylysine Natural products NCC(O)CCC(N)C(O)=O YSMODUONRAFBET-UHFFFAOYSA-N 0.000 description 1
- 230000001627 detrimental effect Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 230000008034 disappearance Effects 0.000 description 1
- 230000006806 disease prevention Effects 0.000 description 1
- PMMYEEVYMWASQN-UHFFFAOYSA-N dl-hydroxyproline Natural products OC1C[NH2+]C(C([O-])=O)C1 PMMYEEVYMWASQN-UHFFFAOYSA-N 0.000 description 1
- 238000001647 drug administration Methods 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- YSMODUONRAFBET-UHNVWZDZSA-N erythro-5-hydroxy-L-lysine Chemical group NC[C@H](O)CC[C@H](N)C(O)=O YSMODUONRAFBET-UHNVWZDZSA-N 0.000 description 1
- 235000020776 essential amino acid Nutrition 0.000 description 1
- 239000003797 essential amino acid Substances 0.000 description 1
- 230000017188 evasion or tolerance of host immune response Effects 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 210000004700 fetal blood Anatomy 0.000 description 1
- 229940012952 fibrinogen Drugs 0.000 description 1
- 239000000834 fixative Substances 0.000 description 1
- 229960000390 fludarabine Drugs 0.000 description 1
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 230000037433 frameshift Effects 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 238000002825 functional assay Methods 0.000 description 1
- 108010074605 gamma-Globulins Proteins 0.000 description 1
- 229940044627 gamma-interferon Drugs 0.000 description 1
- 229960002963 ganciclovir Drugs 0.000 description 1
- IRSCQMHQWWYFCW-UHFFFAOYSA-N ganciclovir Chemical compound O=C1NC(N)=NC2=C1N=CN2COC(CO)CO IRSCQMHQWWYFCW-UHFFFAOYSA-N 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 125000003630 glycyl group Chemical group [H]N([H])C([H])([H])C(*)=O 0.000 description 1
- 210000003714 granulocyte Anatomy 0.000 description 1
- 229910001385 heavy metal Inorganic materials 0.000 description 1
- 208000019691 hematopoietic and lymphoid cell neoplasm Diseases 0.000 description 1
- 208000024200 hematopoietic and lymphoid system neoplasm Diseases 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 239000008241 heterogeneous mixture Substances 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 102000055277 human IL2 Human genes 0.000 description 1
- 102000054751 human RUNX1T1 Human genes 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 125000001165 hydrophobic group Chemical group 0.000 description 1
- 229960002591 hydroxyproline Drugs 0.000 description 1
- 229940121453 idecabtagene vicleucel Drugs 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 230000008004 immune attack Effects 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000008073 immune recognition Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000007813 immunodeficiency Effects 0.000 description 1
- 230000002998 immunogenetic effect Effects 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 230000005917 in vivo anti-tumor Effects 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 210000005007 innate immune system Anatomy 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000005305 interferometry Methods 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 230000003907 kidney function Effects 0.000 description 1
- 238000009533 lab test Methods 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 230000005923 long-lasting effect Effects 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 210000005265 lung cell Anatomy 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 238000002595 magnetic resonance imaging Methods 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 210000003519 mature b lymphocyte Anatomy 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 238000002483 medication Methods 0.000 description 1
- 210000001806 memory b lymphocyte Anatomy 0.000 description 1
- 230000003340 mental effect Effects 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 230000000116 mitigating effect Effects 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical group FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000009826 neoplastic cell growth Effects 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 229960003104 ornithine Drugs 0.000 description 1
- 210000002997 osteoclast Anatomy 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 239000013610 patient sample Substances 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 230000000737 periodic effect Effects 0.000 description 1
- 210000003200 peritoneal cavity Anatomy 0.000 description 1
- 230000002688 persistence Effects 0.000 description 1
- 210000001539 phagocyte Anatomy 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 230000003285 pharmacodynamic effect Effects 0.000 description 1
- 239000002953 phosphate buffered saline Substances 0.000 description 1
- 210000004910 pleural fluid Anatomy 0.000 description 1
- 231100000614 poison Toxicity 0.000 description 1
- 239000002574 poison Substances 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 229940068968 polysorbate 80 Drugs 0.000 description 1
- 238000002600 positron emission tomography Methods 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 230000035935 pregnancy Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000001902 propagating effect Effects 0.000 description 1
- 239000003207 proteasome inhibitor Substances 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000012846 protein folding Effects 0.000 description 1
- 239000012460 protein solution Substances 0.000 description 1
- 229940039716 prothrombin Drugs 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 230000008521 reorganization Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 238000004007 reversed phase HPLC Methods 0.000 description 1
- 231100000279 safety data Toxicity 0.000 description 1
- 238000005070 sampling Methods 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 238000007789 sealing Methods 0.000 description 1
- 230000003248 secreting effect Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 238000004904 shortening Methods 0.000 description 1
- 238000003998 size exclusion chromatography high performance liquid chromatography Methods 0.000 description 1
- 238000001542 size-exclusion chromatography Methods 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 208000024794 sputum Diseases 0.000 description 1
- 210000003802 sputum Anatomy 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 238000011272 standard treatment Methods 0.000 description 1
- 230000003068 static effect Effects 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000012089 stop solution Substances 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 230000002195 synergetic effect Effects 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
- 230000009258 tissue cross reactivity Effects 0.000 description 1
- 230000002110 toxicologic effect Effects 0.000 description 1
- 231100000027 toxicology Toxicity 0.000 description 1
- 231100000041 toxicology testing Toxicity 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 108700012359 toxins Proteins 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 238000003151 transfection method Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 102000035160 transmembrane proteins Human genes 0.000 description 1
- 108091005703 transmembrane proteins Proteins 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
- 229910021642 ultra pure water Inorganic materials 0.000 description 1
- 239000012498 ultrapure water Substances 0.000 description 1
- 231100000402 unacceptable toxicity Toxicity 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 239000011534 wash buffer Substances 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
Images
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
-
- 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
- C07K14/70535—Fc-receptors, e.g. CD16, CD32, CD64 (CD2314/705F)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
- A61K35/17—Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
-
- 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
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- 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
-
- 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
- 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
- C07K14/70578—NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
-
- 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
- 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
- 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
-
- 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
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/31—Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/54—F(ab')2
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/55—Fab or Fab'
-
- 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
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
- C07K2317/732—Antibody-dependent cellular cytotoxicity [ADCC]
-
- 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
- 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
Definitions
- the present invention generally relates to the fields of antibody engineering and cellular immunology, in particular, the present invention relates to an antibody specifically binding to a B-cell maturation antigen (BCMA) comprising a P329G mutation and engineered to express a molecular switch A combination of regulatory chimeric antigen receptor immune effector cells (eg, T cells, NK cells), and relates to the use of said combination in the treatment of diseases associated with the expression of BCMA, such as cancers expressing or overexpressing BCMA.
- BCMA B-cell maturation antigen
- a combination of regulatory chimeric antigen receptor immune effector cells eg, T cells, NK cells
- BCMA B cell maturation antigen
- CD269 CD269, TNFRSF17
- TNFRSF tumor necrosis factor receptor superfamily
- BCMA is a type III transmembrane protein with a cysteine-rich domain (CRD) characteristic of TNFR family members in the extracellular domain (ECD), which forms a ligand-binding motif.
- Ligands of BCMA include B-cell activating factor (BAFF) and B-cell proliferation-inducing ligand (APRIL), wherein B-cell proliferation-inducing ligand (APRIL) binds to BCMA with a higher affinity to promote tumor cell proliferation.
- BAFF B-cell activating factor
- APRIL B-cell proliferation-inducing ligand
- BCMA is mainly expressed on the surface of mature B cells, that is, plasma cells, and is not expressed in normal hematopoietic stem cells and non-blood-derived tissues. BCMA signaling is indispensable for the survival of long-lasting bone marrow plasma cells, but it is not necessary for overall B cell homeostasis. BCMA on the membrane surface can be cleaved by ⁇ -secretase and shed, and the resulting soluble BCMA (sBCMA) may reduce BCMA signal transduction on the membrane surface by blocking BAFF/APRIL ligand binding.
- BCMA was found to be overexpressed in multiple myeloma (Multiple Myeloma, MM) cells, which up-regulates canonical and non-canonical NF- ⁇ B signaling, promotes the growth, survival, and adhesion of MM cells, and induces osteoclasts BCMA expression has become an important marker for the diagnosis of MM.
- MM Multiple Myeloma
- sBCMA in the serum of MM patients increases, which is proportional to the number of MM cells in the bone marrow, and its concentration changes are closely related to the prognosis and treatment response of MM.
- BCMA has become a popular target for the treatment of MM.
- targeted drugs including chimeric antigen receptor T cells.
- CAR-T CAR-T immunotherapy, among which Abecma (idecabtagene vicleucel, ide-cel) of Bluebird (Bluebird) has been approved by the FDA in March 2021, for the treatment of relapsed and refractory MM (RRMM) with 4 lines or above
- RRMM refractory MM
- Ciltacabtagene autoleucel Cilta-cel
- BLA Biologics License Application
- Abecma as the first CAR-T cell therapy targeting BCMA, recognizes and binds to the BCMA protein on multiple myeloma cancer cells, leading to the death of cancer cells expressing BCMA protein. That is, it directly targets the surface antigen BCMA protein of tumor cells through the chimeric antigen receptor (CAR) on CAR-T cells, so as to achieve the purpose of recognizing and killing tumor cells.
- CAR chimeric antigen receptor
- CRS cytokine release syndrome
- NT neurotoxicity
- TAAs tumor-associated antigens
- TSAs tumor-specific antigens
- CD19-targeting CAR-T cells are clinically used to treat CD19-positive blood tumors. In addition to eliminating tumor cells, CD19 CAR-T cells also kill normal B cells.
- CAR-T cells persist in the body, although they have long-term recurrence-free survival with patients It is closely related, but it also leads to the long-term dysplasia of B cells in the body, the loss of humoral immunity, and it is easy to cause infection. Clinical trials have shown that 27-36% of lymphoma patients developed bacterial infection within 30 days after receiving CD19CAR-T cell therapy, and 9.2-28% of patients developed viral infection one month later, requiring a median time of 6.7 months for B cells to recover, 31 -64% of patients required gamma globulin replacement therapy. In addition, the continuous activation of CAR-T cells in the body can easily lead to functional exhaustion, impair its anti-tumor effect, reduce its persistence in the body, and thus reduce the long-term therapeutic effect.
- CAR-T cells In order to reduce the "on-target/off tumor” toxicity of CAR-T cells, the following strategies are generally adopted in the prior art.
- One strategy is to design the antigen-binding domain contained in the CAR to target antigens that are highly expressed on the surface of tumors but not or lowly expressed in normal tissues during CAR design.
- Another strategy is to strictly control the dose of T cells administered, because too many CAR-T cells will increase exponentially after antigen stimulation, which is more likely to cause on-target/off-tumor effects.
- Another strategy is to introduce an inducible suicide gene, such as an inducible Caspase-9 (iCasp9) suicide gene, when constructing a CAR, and administer AP1903 (a gene that can activate iCasp9) when on-target/off-tumor toxicity is observed in the patient.
- an inducible suicide gene such as an inducible Caspase-9 (iCasp9) suicide gene
- AP1903 a gene that can activate iCasp9 when on-target/off-tumor toxicity is observed in the patient.
- Dimerization chemical inducer induces CAR-T cell apoptosis and reduces toxicity
- Zhang Huihui et al. Suicide gene as a "safety switch” to control the preclinical study of CAR-T cell toxicity, Chinese Journal of Cancer Biotherapy, 2021,28(3):225-231
- HSV-TK herpes simplex virus thymidine kinase
- T cells transiently express CAR by electroporation and transfection, and play a therapeutic role through transient killing function
- CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia [J] Leukemia. 2015;29(8):1637–1647).
- the above-mentioned regulatory methods are all carried out after toxicity occurs, and the mitigation effect of toxicity depends on the effect and efficiency of the drug to clear CAR-T cells, and CAR-T cells cannot be recovered after clearing, which affects the long-term anti-tumor efficacy.
- the inventors designed and constructed P329G CAR-T cells targeting BCMA by binding to the Fc domain of an antibody that specifically binds to BCMA molecules containing a P329G mutation, and verified its specificity and controllability in vivo and in vitro. antitumor effect.
- the drug combination of the present invention includes two components: BCMA-specific P329G antibody and P329G CAR-T cells.
- the P329G CAR-T cells are redirected to the tumor cells by recognizing the Fc domain of the P329G antibody, producing tumor recognition and killing effects (see Figure 1B).
- the P329G antibody serves as a bridge connecting P329G CAR-T cells and tumor cells, and acts as a "molecular switch" to regulate the activity of P329G CAR-T cells.
- the present invention provides a P329G mutant antibody capable of specifically binding to BCMA molecules as a "molecular switch" for immune effector cells (for example, T cells, NK cells) expressing CAR polypeptides, including but not Limited to ADI-38497 PG Ab (also referred to herein as “ADI-38497 PG Antibody”, “38497 PG Ab”, “ADI-38497 PG IgG”, “38497 PG IgG”, “PG 38497 Antibody”), ADI-38484 PG Ab (also referred to herein as "ADI-38484 PG antibody”, “38484 PG Ab”, "ADI-38484 PG IgG”, “38484 PG IgG", “PG 38484 antibody”).
- ADI-38497 PG Ab also referred to herein as "ADI-38497 PG Antibody”
- 38497 PG Ab also referred to herein as "ADI-38497 PG Antibody
- ADI-38497 PG IgG also referred to herein
- the present invention obtains an antibody or antigen-binding fragment that specifically binds to a BCMA molecule comprising a heavy chain variable region and a light chain variable region, wherein:
- the heavy chain variable region comprises CDR H1 shown in the amino acid sequence SSSYYWT (SEQ ID NO: 25) according to Kabat numbering, or no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H1 CDR H2 shown in the amino acid sequence SISIAGSTYYNPSLKS (SEQ ID NO: 26), or a variant of no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H2; and the amino acid sequence DRGDQILDV (SEQ ID NO:27) shown in CDR H3, or the variant of no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H3;
- the light chain variable region comprises the amino acid sequence RASQSISRYLN (SEQ ID NO:28) shown in the CDR L1, or the CDR L1 variants with no more than 2 amino acid changes or no more than 1 amino acid change; CDR L2, Or a variant of no more than 2 amino acid changes or no more than 1 amino acid change of the CDR L2; and
- the heavy chain variable region comprises CDR H1 shown in the amino acid sequence NDVIS (SEQ ID NO: 31) according to Kabat numbering, or no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H1 CDR H2 shown in amino acid sequence VIIPIFGIANYAQKFQG (SEQ ID NO: 32), or a variant of no more than 2 amino acid changes or no more than 1 amino acid change of said CDR H2; and amino acid sequence GRGYYSSWLHDI (SEQ ID NO:33) shown in CDR H3, or the variant of no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H3;
- the light chain variable region comprises the amino acid sequence QASQDITNYLN (SEQ CDR L1 shown in ID NO:34), or a variant of no more than 2 amino acid changes or no more than 1 amino acid change of said CDR L1; CDR L2 shown in the amino acid sequence DASNLET (SEQ ID NO:35), Or a variant of no more than 2
- amino acid changes are additions, deletions or substitutions of amino acids.
- the invention provides an antibody or antigen-binding fragment that specifically binds a BCMA molecule comprising a heavy chain variable region and a light chain variable region, wherein:
- the heavy chain variable region comprises CDR H1 shown in the amino acid sequence SSSYYWT (SEQ ID NO:25) according to Kabat numbering; CDR H2 shown in the amino acid sequence SISIAGSTYYNPSLKS (SEQ ID NO:26); and the amino acid sequence The CDR H3 shown in DRGDQILDV (SEQ ID NO:27);
- the light chain variable region comprises the CDR L1 shown in the amino acid sequence RASQSISRYLN (SEQ ID NO:28) according to Kabat numbering; The amino acid sequence AASSLQS (SEQ ID NO: CDR L2 shown in 29); and CDR L3 shown in the amino acid sequence QQKYFDIT (SEQ ID NO:30);
- the heavy chain variable region comprises CDR H1 shown in the amino acid sequence NDVIS (SEQ ID NO:31) according to Kabat numbering; CDR H2 shown in the amino acid sequence VIIPIFGIANYAQKFQG (SEQ ID NO:32); and the amino acid sequence CDR H3 shown in GRGYYSSWLHDI (SEQ ID NO:33);
- the light chain variable region comprises CDR L1 shown in the amino acid sequence QASQDITNYLN (SEQ ID NO:34) according to Kabat numbering; Amino acid sequence DASNLET (SEQ ID NO: CDR L2 shown in 35); and CDR L3 shown in the amino acid sequence QQAFDLIT (SEQ ID NO:36).
- the present invention obtains an antibody or antigen-binding fragment that specifically binds to a BCMA molecule comprising a heavy chain variable region and a light chain variable region, wherein:
- the heavy chain variable region comprises the sequence of SEQ ID NO: 2 or is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical thereto sequence
- the light chain variable region comprises the sequence of SEQ ID NO: 3 or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% thereof % identity sequence
- the heavy chain variable region comprises the sequence of SEQ ID NO: 9 or is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical thereto sequence
- the light chain variable region comprises the sequence of SEQ ID NO: 10 or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% thereof % identity sequence.
- the present invention obtains an antibody or antigen-binding fragment that specifically binds to a BCMA molecule comprising a heavy chain variable region and a light chain variable region, wherein:
- the heavy chain variable region comprises the sequence of SEQ ID NO:2, and the light chain variable region comprises the sequence of SEQ ID NO:3;
- the heavy chain variable region comprises the sequence of SEQ ID NO:9, and the light chain variable region comprises the sequence of SEQ ID NO:10.
- an antibody of the invention that specifically binds a BCMA molecule is an IgG1, IgG2, IgG3 or IgG4 antibody; preferably, it is an IgG1 or IgG4 antibody; more preferably, it is an IgG1 antibody.
- the antigen-binding fragment of an antibody of the invention that specifically binds a BCMA molecule is Fab, Fab', F(ab') 2 , Fv, single chain Fv, single chain Fab, diabody.
- an antibody with a mutated Fc domain is obtained by mutating the amino acid at the P329 position according to EU numbering to glycine (G), wherein, with the unmutated parental antibody The Fc ⁇ receptor binding of the mutant Fc domain was reduced compared to the Fc ⁇ receptor binding of the Fc domain.
- the mutant Fc domain is a mutant Fc domain of an IgG1, IgG2, IgG3 or IgG4 antibody, preferably, the mutant Fc domain is a mutant Fc domain of an IgG1 or IgG4 antibody; more preferably , the mutant Fc domain is a mutant Fc domain of an IgG1 antibody;
- the antibody or antigen-binding fragment that specifically binds to a BCMA molecule comprises the heavy chain constant region sequence shown in SEQ ID NO: 5 or at least 90%, 91%, 92%, 93%, 94%, A sequence of 95%, 96%, 97%, 98% or 99% identity and wherein the amino acid at position P329 according to EU numbering is mutated to G;
- the antibody or antigen-binding fragment comprises or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% of the heavy chain constant region sequence shown in SEQ ID NO: 5 , a sequence of 98% or 99% identity and wherein the amino acid at position P329 according to EU numbering is mutated to G; and the light chain constant region sequence shown in SEQ ID NO: 6 or at least 90%, 91%, 92% therewith %, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical sequences.
- the antibody or antigen-binding fragment that specifically binds to a BCMA molecule comprises the heavy chain constant region sequence shown in SEQ ID NO:5 and the light chain constant region sequence shown in SEQ ID NO:6.
- the present invention provides a nucleic acid encoding the antibody of the first aspect of the invention, a vector comprising the nucleic acid encoding the antibody, a cell comprising the nucleic acid molecule or vector, and a method for producing the antibody, the The method includes culturing the antibody or antigen-binding fragment encoding the antibody or antigen-binding fragment encoding the BCMA molecule described in the first aspect of the first aspect of the present invention that is introduced under conditions suitable for expressing the nucleic acid encoding the antibody or antigen-binding fragment that specifically binds to the BCMA molecule described in the first aspect of the present invention.
- the host cell is prokaryotic or eukaryotic, more preferably selected from Escherichia coli cells, yeast cells, mammalian cells or other cells suitable for preparing antibodies or antigen-binding fragments thereof, most preferably, the Host cells are HEK293 cells or CHO cells.
- the present invention provides a pharmaceutical combination comprising
- a first component selected from immune effector cells (for example, T cells, NK cells) expressing a molecular switch-regulated CAR polypeptide, a nucleic acid molecule encoding the CAR polypeptide, a vector comprising the nucleic acid molecule, and any combination of them; and
- immune effector cells for example, T cells, NK cells
- the second component which is an antibody or antigen-binding fragment (also known as a P329G mutant antibody) that specifically binds to a BCMA molecule comprising a P329G mutation, for example, the P329G mutant antibody of the first aspect of the present invention
- the molecular switch-regulated CAR polypeptide comprises
- scFv sequence comprises the following sequence that can specifically bind to the Fc domain of an antibody comprising a P329G mutation, but cannot specifically bind to the Fc domain of an unmutated parental antibody:
- CDR L light chain complementarity determining region 1 shown in the amino acid sequence RSSTGAVTTSNYAN (SEQ ID NO: 22), or a variant of said CDR L1 with no more than 2 amino acid changes or no more than 1 amino acid change;
- amino acid change is an addition, deletion or substitution of an amino acid
- n is an integer from 1 to 10, such as an integer from 1 to 4; for example SEQ ID NO: 14 the sequence shown;
- CD8 ⁇ hinge region or variants thereof with 1-5 amino acid modifications for example, the sequence shown in SEQ ID NO: 18 or variants thereof with 1-2 amino acid modifications;
- Transmembrane region which is selected from CD8 transmembrane domain or variants with 1-5 amino acid modifications, for example, the sequence shown in SEQ ID NO: 15 or with 1-2 amino acids Modified variants;
- Costimulatory signaling domain which is selected from 4-1BB costimulatory domain or variants with 1-5 amino acid modifications, for example, the sequence shown in SEQ ID NO: 16 or its variant with 1 - 2 amino acid modified variants;
- Stimulatory signaling domain which is a CD3 ⁇ signaling domain or a variant having 1-10 amino acid modifications, for example, the sequence shown in SEQ ID NO: 17 or having 1-10, 1 - 5 amino acid modified variants;
- amino acid modification is addition, deletion or substitution of amino acid.
- the molecular switch-regulated CAR polypeptide described in the pharmaceutical combination of the present invention comprises
- scFv sequence comprises the following sequence that can specifically bind to the Fc domain of an antibody comprising a P329G mutation, but cannot specifically bind to the Fc domain of an unmutated parental antibody:
- a heavy chain variable region comprising or having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% of the sequence of SEQ ID NO: 12 or 99% identical sequences, and
- transmembrane region which is selected from the CD8 transmembrane domain shown in SEQ ID NO: 15 or a variant with 1 amino acid modification;
- co-stimulatory signaling domain which is selected from the 4-1BB co-stimulatory domain shown in SEQ ID NO: 16 or a variant with 1 amino acid modification;
- Stimulatory signaling domain which is selected from the CD3 ⁇ signaling domain shown in SEQ ID NO: 17 or a variant with 1 amino acid modification;
- amino acid modification is addition, deletion or substitution of amino acid.
- the molecular switch-regulated CAR polypeptide described in the pharmaceutical combination of the present invention further comprises a signal peptide sequence at the N-terminus, for example, the signal peptide sequence shown in SEQ ID NO: 11,
- the molecular switch-regulated CAR polypeptide in the pharmaceutical combination of the present invention has the amino acid sequence shown in SEQ ID NO: 1 or at least 90%, 91%, 92%, 93%, 94%, Sequences that are 95%, 96%, 97%, 98% or 99% identical.
- the present invention provides a nucleic acid encoding the molecular switch-regulated CAR polypeptide described in the pharmaceutical combination of the present invention, a vector comprising the nucleic acid encoding the CAR polypeptide, and a cell comprising the CAR nucleic acid molecule or vector, Or a cell expressing the CAR polypeptide, preferably, the cell is an autologous T cell or an allogeneic T cell.
- the nucleic acid molecule encoding the molecular switch-regulated CAR polypeptide described in the pharmaceutical combination of the present invention encodes the amino acid sequence shown in SEQ ID NO: 1 or has at least 90%, 91%, 92%, 93 A nucleic acid molecule having an amino acid sequence of %, 94%, 95%, 96%, 97%, 98% or 99% identity.
- the vector comprising the nucleic acid molecule encoding the molecular switch-regulated CAR polypeptide described in the pharmaceutical combination of the present invention is selected from DNA vectors, RNA vectors, plasmids, lentiviral vectors, adenoviral vectors or retroviral vectors.
- the immune effector cells in the pharmaceutical combination of the present invention are autologous T cells, NK cells or allogeneic T cells, NK cells that express the molecular switch-regulated CAR polypeptide of the present invention.
- Cells for example, the immune effector cells are T cells isolated from human peripheral blood mononuclear cells (PBMC), T cells prepared from NK cells expressing the molecular switch-regulated CAR polypeptide of the present invention, NK cells.
- PBMC peripheral blood mononuclear cells
- the present invention utilizes primary P329G CAR-T cells prepared from human PBMC derived from multiple different donors, and utilizes a P329G mutated anti-BCMA humanized antibody to evaluate P329G in an in vitro co-culture system.
- CAR-T cells target BCMA-expressing tumor cells through the effector function of antibodies. Therefore, the P329G mutated anti-BCMA antibody can be used as a "molecular switch" to regulate the recognition and killing activity of P329G CAR-T cells on BCMA-positive tumor cells.
- the effector function in vitro is comparable to that of traditional CAR-T cells directly targeting BCMA-positive tumor cells, but the activity of traditional CAR-T cells does not depend on the P329G mutant antibody.
- the present invention verifies the anti-tumor effect of P329G CAR-T cells combined with P329G mutant antibody in immunodeficient mice inoculated with BCMA-positive human tumor cell line-derived tumors, and the dose of the antibody, interval etc. were studied.
- the immune effector cells expressing the CAR polypeptide of the present invention are injected at a concentration of 1 ⁇ 10 6 cells/kg body weight to 10 ⁇ 10 6 cells/kg body weight, for example, 1 ⁇ 10 6 cells/kg body weight. 10 6 cells/kg body weight, 2 ⁇ 10 6 cells/kg body weight, 3 ⁇ 10 6 cells/kg body weight, 5 ⁇ 10 6 cells/kg body weight, 7 ⁇ 10 6 cells/kg body weight, 9 ⁇ A dose of 106 cells/kg body weight, 10 x 106 cells/kg body weight is administered intravenously to the subject in single or multiple doses; and
- the P329G mutant antibody described in the pharmaceutical combination of the present invention is dosed at 0.1-10 mg/kg, preferably 0.1 mg/kg, 0.3 mg/kg, 0.5 mg/kg, 1 mg/kg, 3 mg/kg, 5 mg/kg kg, 7 mg/kg, 9 mg/kg, 10 mg/kg dosage unit form, preferably parenterally, more preferably intravenously administered to the subject.
- (i) and (ii) in the pharmaceutical combination of the present invention are administered separately, simultaneously or sequentially, for example, administering (ii) on the first day, administering (i) intravenously on the same day, and then administering (i) at a certain frequency multiple administrations of (ii), while determining whether to administer (i) multiple times by monitoring the in vivo PK concentration of (i) and the desired therapeutic efficacy endpoint; or
- the present invention provides the use of the pharmaceutical combination of the present invention for treating BCMA-related diseases in a subject, comprising administering to the subject a therapeutically effective amount of the pharmaceutical combination defined in the aforementioned third aspect , wherein the BCMA-related disease is, for example, a cancer that expresses or overexpresses BCMA, such as relapsed/refractory multiple myeloma (relapsed/refractory multiple myeloma, RRMM).
- the BCMA-related disease is, for example, a cancer that expresses or overexpresses BCMA, such as relapsed/refractory multiple myeloma (relapsed/refractory multiple myeloma, RRMM).
- the present invention provides the use of the pharmaceutical combination of the present invention in the preparation of a medicament for the treatment of BCMA-related diseases, such as cancers expressing or overexpressing BCMA, said cancers
- BCMA-related diseases such as cancers expressing or overexpressing BCMA
- RRMM relapsed/refractory multiple myeloma
- the present invention provides a method for treating a disease associated with BCMA, the method comprising administering to a subject a therapeutically effective amount of the pharmaceutical combination of the present invention, the disease being, for example, expressing or overexpressing BCMA Cancer, such as relapsed/refractory multiple myeloma (RRMM).
- a disease associated with BCMA the method comprising administering to a subject a therapeutically effective amount of the pharmaceutical combination of the present invention, the disease being, for example, expressing or overexpressing BCMA Cancer, such as relapsed/refractory multiple myeloma (RRMM).
- RRMM relapsed/refractory multiple myeloma
- the present invention provides a kit of parts comprising the pharmaceutical combination as defined in the aforementioned third aspect, preferably said kit is in the form of a dosage unit of the drugs.
- the present invention provides a drug complex, which is a compound composed of
- immune effector cells eg, T cells, NK cells
- an antibody or antigen-binding fragment also known as a P329G mutant antibody
- a BCMA molecule comprising a P329G mutation
- the immune effector cells are T cells expressing the molecular switch regulatory CAR polypeptide described in the pharmaceutical combination of the present invention prepared from autologous T cells or allogeneic T cells
- the immune effector cells are from human T cells expressing the molecular switch regulatory CAR polypeptide of the present invention prepared from T cells isolated from PBMC;
- the P329G mutant antibody is ADI-38497 PG Ab and/or ADI-38484 PG Ab.
- the present invention also provides the use of the drug complex for treating a BCMA-related disease in a subject, preferably, the BCMA-related disease is, for example, a cancer expressing or overexpressing BCMA, the cancer An example is relapsed/refractory multiple myeloma (RRMM).
- the BCMA-related disease is, for example, a cancer expressing or overexpressing BCMA, the cancer
- RRMM relapsed/refractory multiple myeloma
- the present invention obtains a high-affinity BCMA-specific P329G antibody through in vitro binding ability, affinity and Fc effector function detection, and the antibody can simultaneously bind BCMA antigen and P329G CAR molecule to exert a bridging effect.
- the present invention constructs an in vitro co-culture system by using the constructed P329G CAR structural molecule and combining the P329G CAR-T cells prepared by the CAR molecule with a BCMA-specific P329G antibody, and then co-culturing with BCMA-positive MM cells in vitro , in this system, it was verified that P329G antibody acts as a "molecular switch" to regulate the effect of P329G CAR-T cell activity, that is, only in the presence of P329G mutant antibody, P329G CAR-T cells can be activated, proliferate, and secrete effector cells Factors and killing effects, and these effects are P329G antibody dose-dependent, with the increase of antibody dose, P329G CAR-T cell recognition and killing effects are enhanced.
- the WT antibody without the P329G mutation could not stimulate the effector function of P329G CAR-T cells.
- the in vitro experiments of the present invention show that the soluble BCMA antigen does not affect the activity of P329G CAR-T cells when used in combination with BCMA-specific P329G antibodies, while the soluble BCMA antigen has a significant inhibitory effect on traditional CAR-T cells.
- the P329G CAR-T cells of the present invention combined with P329G antibodies produced good anti-tumor effects, and the anti-tumor effects were at least as good as those of traditional CAR-T cells.
- Figure 1A shows the expression of CAR in CD3 + cells, CD4 + , and CD8 + T cell subsets after T cells were transduced with the HuR968B and Blue21 CARs constructed in Example 1-1.
- Figure 1B shows the mechanism of action of P329G CAR-T cells targeting BCMA-expressing target cells mediated by P329G antibody.
- SP signal peptide
- TMD transmembrane domain
- ICD intracellular domain
- CSD co-stimulatory signal domain ( costimulatory domain)
- SSD stimulating signaling domain.
- the extracellular domain comprises an antigen-binding part capable of specifically binding to a mutant Fc domain containing a P329G mutation, and the antigen-binding part comprises a heavy chain variable region (VH) and Light Chain Variable Region (VHL).
- VH heavy chain variable region
- VHL Light Chain Variable Region
- Fig. 2A shows a schematic diagram of a method for measuring antibody affinity by surface plasmon resonance (SPR).
- Figure 2B shows representative affinity profiles of ADI-38497 PG antibody to recombinant human, cynomolgus monkey, mouse, rat and rabbit BCMA proteins measured by SPR.
- Figure 2C shows the binding ability of P329G BCMA antibody to CHO-GS cells stably expressing human, cynomolgus and mouse BCMA.
- Figure 2D shows the binding activity of P329G BCMA antibody to BCMA-positive multiple myeloma cell lines MM.1s, RPMI8226, U266, H929, L363 and AMO1.
- Figure 3A shows a schematic diagram of the detection of the affinity of the specific single-chain antibody against the P329G mutation-rabbit Fc fusion protein and the ADI-38497 P329G mutation antibody by surface plasmon resonance (SPR).
- SPR surface plasmon resonance
- Figure 3B shows representative affinity profiles of ADI-38497 PG antibody and wild-type antibody binding to anti-PG scFv fusion protein by SPR.
- Fig. 3C shows a schematic diagram of the method for measuring antibody affinity (Avidity) by surface plasmon resonance (SPR).
- Figure 3D shows representative affinity profiles of ADI-38497 PG antibody and wild-type antibody and anti-PG scFv fusion protein (interim, P329G CAR extracellular domain contains anti-PG scFv) by SPR.
- Figure 3E shows the binding ability of ADI-38497WT antibody and ADI-38497 PG antibody to P329G CAR-T cells.
- Figure 4A shows the ability of ADI-38497WT antibody and ADI-38497 PG antibody to mediate ADCC killing.
- Figure 4B shows the ability of ADI-38497WT antibody and ADI-38497 PG antibody to mediate ADCP killing.
- Figure 4C shows the ability of the ADI-38497 PG antibody to mediate lysis of target cells.
- Figure 5A shows that for H929 cells, the ADI-38497 PG antibody containing the P329G mutation specifically mediates the activation of CAR + -T.
- Figure 5B shows that different BCMA antibodies (ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, GSK PG IgG) induced the activation effect of L363 target cells on HuR968B CAR-T cells.
- BCMA antibodies ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, GSK PG IgG
- Figure 5C shows the proliferation of HuR968B CAR-T cells stimulated by coated ADI-38497WT antibody or ADI-38497 PG antibody.
- Figure 5D shows that HuR968B CAR-T cells were co-cultured with H929 cells and RPMI8226 cells. After adding different concentrations of BCMA antibodies (ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or GSK PG IgG as a positive control ) after the release of effector cytokines secreted by CAR-T cells.
- BCMA antibodies ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or GSK PG IgG as a positive control
- FIG. 5E shows that HuR968B CAR-T cells were co-cultured with different tumor cells. After adding different concentrations of BCMA antibodies (ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or GSK PG IgG), CAR-T cells The release of secreted effector cytokines results.
- BCMA antibodies ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or GSK PG IgG
- FIG. 5F shows that different BCMA antibodies (ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or GSK PG IgG) induced HuR968B CAR-T cells to tumor cells with different BCMA expression levels (H929 ++ cells, RPMI8226 +++ cells, AMO1 + cells and L363 + cells).
- BCMA antibodies ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or GSK PG IgG
- Figure 5G shows the killing effect of P329G BCMA antibody-induced CAR-T production on tumor cells.
- Figure 6 shows the effect of different concentrations of free BCMA protein on the killing effect of HuR968B CAR-T and Blue21CAR-T cells.
- Figure 7A and Figure 7B show the results of pharmacokinetic experiments of ADI-38497 PG antibody in mice.
- FIG 8A shows the therapeutic effect of different doses of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 high-expressing BCMA tumor cells.
- cCAR-T means a conventional CAR-T, ie, Blue21CAR-T.
- Figure 8B shows the body weight changes of immunodeficient tumor-bearing mice inoculated subcutaneously with different doses of PG antibody combined with PG CAR-T cells in human H929 high-expressing BCMA tumor cells.
- Figure 8C shows the expansion of PG CAR-T cells in mice when different doses of PG antibody combined with PG CAR-T cells were inoculated subcutaneously in immunodeficient tumor-bearing mice with human H929 high-expressing BCMA tumor cells.
- FIG. 9A shows the therapeutic effect of different doses of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human L363 low-expression BCMA tumor cells.
- cCAR-T means conventional CAR-T, ie, Blue21CAR-T.
- Figure 9B shows the body weight changes of immunodeficient tumor-bearing mice inoculated subcutaneously with different doses of PG antibody combined with PG CAR-T cells in human L363 low-expressing BCMA tumor cells.
- Figure 9C shows the expansion of PG CAR-T cells in mice when different doses of PG antibody combined with PG CAR-T cells were inoculated subcutaneously in immunodeficient tumor-bearing mice with low expression of human L363 BCMA tumor cells.
- Figure 10A shows the therapeutic effect of PG antibody combined with different doses of PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- Figure 10B shows the expansion of PG CAR-T cells in mice when PG antibodies were combined with different doses of PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- Fig. 11A shows the frequency of administration of the PG antibody in the experiment of Example 11-1.
- Figure 11B shows the therapeutic effect of different administration frequencies of PG antibody when combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- Figure 11C shows the expansion of PG CAR-T cells in mice in the experiment of Example 11-1.
- Figure 12A shows the fluorescence images of the therapeutic effects of different doses of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated with human H929-luc tumor cells in the tail vein.
- Figure 12B shows the therapeutic effect of different doses of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated with human H929-luc tumor cells in the tail vein.
- Figure 12C shows the body weight changes of immunodeficient tumor-bearing mice inoculated with human H929-luc tumor cells in the tail vein of different doses of PG antibody combined with PG CAR-T cells.
- Figure 13A shows the therapeutic effect of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- Figure 13B shows the changes in body weight of the mice treated with PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- Figure 13C and Figure 13D show the hematology and blood biochemical detection results of the mice treated with PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- BCMA and “B cell maturation antigen” are used interchangeably and include variants, isoforms, species homologs and analogs of human BCMA having at least one epitope identical to BCMA (e.g., human BCMA) .
- BCMA proteins may also include fragments of BCMA, such as the extracellular domain and fragments of the extracellular domain, eg, fragments that retain the ability to bind to any antibody of the invention.
- BCMA antibody As used herein, the terms “BCMA antibody”, “antibody against BCMA”, “antibody that specifically binds BCMA”, “antibody that specifically targets BCMA”, “antibody that specifically recognizes BCMA” are used interchangeably, Means an antibody capable of specifically binding to B-cell maturation antigen (BCMA).
- antibody is used herein in the broadest sense to refer to a protein comprising an antigen binding site, encompassing natural and artificial antibodies of various structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (for example, bispecific antibodies), single chain antibodies, whole antibodies and antibody fragments.
- the antibodies of the invention are single domain antibodies or heavy chain antibodies.
- Antibody fragment or "antigen-binding fragment” are used interchangeably herein to refer to a molecule, other than an intact antibody, that comprises a portion of an intact antibody and that binds the antigen to which the intact antibody binds.
- antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, Fv, single chain Fv, single chain Fab, diabody.
- scFv refers to a fusion protein comprising at least one antibody fragment comprising a light chain variable region and at least one antibody fragment comprising a heavy chain variable region, wherein the light chain variable region and the heavy chain variable region are optionally
- the scFvs are continuously linked by a flexible short polypeptide linker and can be expressed as a single chain polypeptide in which the scFv retains the specificity of the intact antibody from which it was derived.
- a scFv can have a VL variable region and a VH variable region in any order (e.g., relative to the N-terminus and C-terminus of a polypeptide), a scFv can comprise a VL-linker-VH or can comprise VH-Joint-VL.
- a “complementarity determining region” or “CDR region” or “CDR” or “hypervariable region” is an antibody variable domain that is hypervariable in sequence and forms a structurally defined loop ("hypervariable loop") and/or Regions containing antigen contact residues ("antigen contact points").
- the CDRs are primarily responsible for binding to antigenic epitopes.
- the CDRs of the heavy and light chains are commonly referred to as CDR1, CDR2 and CDR3, numbered sequentially starting from the N-terminus.
- CDR H1, CDR H2, and CDR H3 The CDRs located in the variable domain of the antibody heavy chain are referred to as CDR H1, CDR H2, and CDR H3, while the CDRs located in the variable domain of the antibody light chain are referred to as CDR L1, CDR L2, and CDR L3.
- each CDR can be determined using any one or combination of a number of well-known antibody CDR assignment systems, including For example: Chothia based on the three-dimensional structure of antibodies and the topology of CDR loops (Chothia et al., (1989) Nature 342:877-883, Al-Lazikani et al., "Standard conformations for the canonical structures of immunoglobulins", Journal of Molecular Biology, 273, 927-948 (1997)), Kabat based on antibody sequence variability (Kabat et al., Sequences of Proteins of Immunological Interest, 4th ed., U.S.
- a CDR can also be determined based on having the same Kabat numbering position as a reference CDR sequence (eg, any of the exemplified CDRs of the present invention).
- a reference CDR sequence eg, any of the exemplified CDRs of the present invention.
- reference is made to numbering positions according to the Kabat numbering system.
- CDRs vary from antibody to antibody, only a limited number of amino acid positions within a CDR are directly involved in antigen binding. Using at least two of the Kabat, Chothia, AbM and Contact methods, the region of minimal overlap can be determined, thereby providing a "minimum binding unit" for antigen binding.
- a minimal binding unit may be a subsection of a CDR.
- the residues of the remainder of the CDR sequences can be determined from the structure and protein folding of the antibody. Accordingly, the invention also contemplates variations of any of the CDRs presented herein. For example, in a variant of a CDR, the amino acid residues of the smallest binding unit can remain unchanged, while the remaining CDR residues defined according to Kabat or Chothia or AbM can be replaced by conservative amino acid residues.
- chimeric antibody is an antibody molecule in which (a) the constant region or part thereof is altered, substituted or exchanged such that the antigen binding site is of a different or altered class and/or species of constant region or confers a chimeric (e.g., enzymes, toxins, hormones, growth factors, drugs), etc.; or (b) replace the variable region or part thereof with a variable region having different or altered antigenic specificity Alter, replace or exchange.
- murine antibodies can be modified by exchanging their constant regions with those from human immunoglobulins. Due to the exchange of human constant regions, the chimeric antibody can retain its specificity in recognizing the antigen while having reduced antigenicity in humans as compared to the original murine antibody.
- a “humanized” antibody refers to a chimeric antibody that comprises amino acid residues from non-human CDRs and amino acid residues from human FRs.
- all or substantially all of the CDRs (eg, CDRs) in the humanized antibody correspond to those of a non-human antibody
- all or substantially all of the FRs correspond to those of a human antibody.
- a humanized antibody optionally can comprise at least a portion of an antibody constant region derived from a human antibody.
- a "humanized form" of an antibody (eg, a non-human antibody) refers to an antibody that has been humanized.
- Human antibody refers to an antibody having an amino acid sequence corresponding to that of an antibody produced by a human or human cell or derived from a non-human source using a human antibody library or other human Antibody coding sequence. This definition of a human antibody specifically excludes humanized antibodies comprising non-human antigen-binding residues.
- Fc region refers to the C-terminal region of an immunoglobulin heavy chain, including native sequence Fc regions and variant Fc regions.
- the Fc region of a human IgG heavy chain is generally defined as the segment from the amino acid residue at Cys226 or Pro230 to the carboxyl-terminus, and the lysine residue at position 447 at the C-terminus of the Fc region (according to the EU numbering system) may be present or absent.
- whole antibody compositions can include a population of antibodies in which all K447 residues have been eliminated, a population of antibodies in which no K447 residue has been eliminated, or a population of antibodies in which antibodies with and without K447 residues are mixed.
- the Fc region of an immunoglobulin comprises two constant domain domains, CH2 and CH3, and in other embodiments, the Fc region of an immunoglobulin comprises three constant domains, CH2, CH3 and CH4.
- Binding of IgG to Fc ⁇ receptors or C1q is dependent on residues localized in the hinge region and CH2 domain. Two regions of the CH2 domain are critical for Fc ⁇ R and complement C1q binding and have unique sequences in IgG2 and IgG4. Substitution of residues 233-236 in human IgG1 and IgG2 and substitution of residues 327, 330 and 331 in human IgG4 has been shown to substantially reduce ADCC and CDC activity (Armour et al., Eur. J. Immunol. 29(8 ), 1999, 2613-2624; Shields et al., J. Biol. Chem. 276(9), 2001, 6591-6604).
- Functional Fc region and “functional Fc region” and similar terms may be used interchangeably, referring to an Fc region having effector functions of a wild-type Fc region.
- Variant Fc region “Fc mutant”, “Fc region carrying a mutation”, “mutant Fc region”, “Fc region variant”, “Fc variant”, “variant Fc region” and “mutated Fc region” Similar terms such as “region” can be used interchangeably, and refer to an Fc region comprising at least one amino acid modification that is distinguished from a native sequence Fc region/wild-type Fc region.
- a variant Fc region comprises an amino acid sequence that differs from the amino acid sequence of a native sequence Fc region by one or more amino acid substitutions, deletions or additions.
- the variant Fc region has at least one amino acid substitution compared to the Fc region of wild-type IgG, the at least one amino acid substitution being a glycine (G) substitution of the amino acid at position P329 according to EU numbering.
- Fc receptor refers to a molecule that binds the Fc region of an antibody.
- the FcR is a native human FcR.
- the FcR is a receptor that binds an IgG antibody, namely an Fc ⁇ R, including three receptors, Fc ⁇ RI (CD64), Fc ⁇ RII (CD32), and Fc ⁇ RIII (CD16), and allelic variants and variants of these receptors. splice form.
- Fc ⁇ RII receptors include Fc ⁇ RIIA and Fc ⁇ RIIB
- Fc ⁇ RIII receptors include Fc ⁇ RIIIA and Fc ⁇ RIIIB.
- effector functions refers to those biological activities attributable to the Fc region of an immunoglobulin that vary with immunoglobulin isotype.
- immunoglobulin effector functions include: Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), cytokine secretion, immune complex-mediated antigen Uptake of antigen by presenting cells, Clq binding and complement-dependent cytotoxicity (CDC), downregulation of cell surface receptors (eg, B cell receptors), and B cell activation.
- ADCC antibody-dependent cell-mediated cytotoxicity
- NK natural killer cells
- the chimeric antigen receptors of the invention provide antibody-dependent cytotoxicity of T lymphocytes, enhance antibody-dependent cytotoxicity of NK cells.
- the chimeric antigen receptor of the present invention induces the activation, continuous proliferation and exertion of T cells expressing the chimeric antigen receptor by binding to the antibody (or other anti-tumor molecules comprising the Fc part) that binds to the tumor cell Other anti-tumor molecules containing Fc moieties) mediated specific cytotoxicity against cancer cells of interest.
- ADCP antibody-dependent cellular phagocytosis
- complement-dependent cytotoxicity refers to the lysis of target cells in the presence of complement.
- the complement system is part of the innate immune system made up of a series of proteins.
- the proteins of the complement system are called “complement”, represented by abbreviations C1, C2, C3, etc., which are a group of heat-labile proteins that exist in human or vertebrate serum and interstitial fluid, and have enzymatic activity after activation.
- CIq is the first component of the complement-dependent cytotoxicity (CDC) pathway, capable of binding six antibodies, but binding to two IgGs is sufficient to activate the complement cascade.
- Activation of the classical complement pathway is initiated by binding of the first component of the complement system (C1q) to antibodies (of the appropriate subclass) that bind the relevant antigen, activating a series of complement cascades that form holes in the target cell membrane, resulting in target cell death.
- C1q first component of the complement system
- a CDC assay can be performed, for example, by the method described in Gazzano-Santoro et al., J. Immunol. Methods 202:163 (1996).
- variable region refers to the domains of an antibody heavy or light chain that participate in the binding of the antibody to an antigen.
- the variable domains of the heavy and light chains of native antibodies generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three complementarity determining regions (CDRs).
- FRs conserved framework regions
- CDRs complementarity determining regions
- bind or “specifically bind” means that the binding is selective for the antigen and can be distinguished from unwanted or non-specific interactions.
- the ability of an antibody to bind a particular antigen can be determined by enzyme-linked immunosorbent assay (ELISA), SPR or biofilm layer interferometry techniques or other conventional binding assays known in the art.
- stimulation refers to a primary response induced by the binding of a stimulatory molecule (e.g., a TCR/CD3 complex) to its corresponding ligand, which thus mediates a signal transduction event, such as, but not limited to, via a TCR/CD3 complex body signal transduction.
- a stimulatory molecule e.g., a TCR/CD3 complex
- Stimulation can mediate altered expression of certain molecules, such as down-regulation of TGF- ⁇ and/or reorganization of cytoskeletal structures, etc.
- the term "stimulatory molecule” refers to a molecule expressed by a T cell that provides a primary cytoplasmic signaling sequence that modulates TCR complex activation in a stimulatory manner in at least some aspect of the T cell signaling pathway.
- primary activation In one embodiment, primary signals are elicited, eg, by binding of the TCR/CD3 complex to peptide-loaded MHC molecules and result in mediation of T cell responses including, but not limited to, proliferation, activation, differentiation, and the like.
- the intracellular signaling domain in any one or more CARs of the invention comprises an intracellular signaling sequence, eg, the primary signaling sequence of CD3 ⁇ .
- CD3 ⁇ is defined as the protein given by GenBan Accession No. BAG36664.1 or its equivalent
- CD3 ⁇ stimulatory signaling domain is defined as amino acid residues from the cytoplasmic domain of the CD3 ⁇ chain sufficient for functional Transmits the initial signal necessary for T cell activation.
- the cytoplasmic domain of CD3 ⁇ comprises residues 52 to 164 of GenBank accession number BAG36664.1 or as a functional ortholog thereof from a non-human species (e.g., mouse, rodent, equivalent residues for monkeys, apes, etc.).
- the "CD3 ⁇ stimulatory signaling domain” is the sequence provided in SEQ ID NO: 17 or a variant thereof.
- costimulatory molecule refers to a corresponding binding partner on a cell that specifically binds to a costimulatory ligand to mediate a costimulatory response (eg, but not limited to, proliferation) of the cell.
- Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that contribute to an effective immune response.
- Costimulatory molecules include but are not limited to MHC class I molecules, TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocyte activation molecules (SLAM proteins), activating NK cell receptors, OX40 , CD40, GITR, 4-1BB (ie CD137), CD27 and CD28.
- the "co-stimulatory molecule” is 4-1BB (ie, CD137).
- Costimulatory signaling domain refers to the intracellular portion of a costimulatory molecule.
- 4-1BB refers to a member of the TNFR superfamily having the amino acid sequence provided as GenBank Accession No. AAA62478.2 or the equivalent residues from a non-human species (e.g., mouse, rodent, monkey, ape, etc.) and "4-1BB co-stimulatory signaling domain" is defined as amino acid residues 214-255 of GenBank accession number AAA62478.2 or equivalent residues from a non-human species (eg, mouse, rodent, monkey, ape, etc.) .
- the "4-1BB co-stimulatory domain” is the sequence provided as SEQ ID NO: 16 or equivalent residues from a non-human species (e.g., mouse, rodent, monkey, ape, etc.).
- signaling pathway refers to the biochemical relationship between various signaling molecules that play a role in propagating a signal from one part of a cell to another.
- cytokine is a general term for proteins released by one cell population to act as intercellular mediators on another cell.
- cytokines are lymphokines, monokines, interleukins (IL), such as IL-1, IL-1 ⁇ , IL-2, IL-3, IL-4, IL-5, IL-6, IL- 7.
- antibodies of the invention are purified to greater than 95% or 99% purity, such as by, for example, electrophoresis (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatography (e.g., ion exchange or reverse phase HPLC).
- electrophoresis e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
- chromatography e.g., ion exchange or reverse phase HPLC.
- nucleic acid refers to a nucleic acid molecule that has been separated from components of its natural environment.
- An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location other than its natural chromosomal location.
- isolated nucleic acid encoding an antibody of the invention refers to one or more nucleic acid molecules encoding a chain of an antibody of the invention or fragments thereof, including such nucleic acid molecules in a single vector or in separate vectors, and present in a host cell Such nucleic acid molecules at one or more positions in .
- the sequences are aligned for optimal comparison purposes (e.g., a first and second amino acid sequence or nucleic acid sequence may be placed between a first and a second amino acid sequence or nucleic acid sequence for optimal alignment). Gaps may be introduced in one or both or non-homologous sequences may be discarded for comparison purposes).
- the length of the aligned reference sequence is at least 30%, preferably at least 40%, more preferably at least 50%, 60% and even more preferably at least 70%, 80% , 90%, 100% of the reference sequence length.
- the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
- the comparison of sequences and the calculation of percent identity between two sequences can be accomplished using a mathematical algorithm.
- the Needlema and Wunsch ((1970) J. Mol. Biol. 48:444-453) algorithm (available at http://www.gcg.com available), use the Blossum 62 matrix or the PAM250 matrix with gap weights of 16, 14, 12, 10, 8, 6 or 4 and length weights of 1, 2, 3, 4, 5 or 6 to determine the distance between two amino acid sequences. percent identity.
- using the GAP program in the GCG software package (available at http://www.gcg.com), using the NWSgapdna.CMP matrix and gap weights of 40, 50, 60, 70 or 80 and Length weights of 1, 2, 3, 4, 5 or 6 determine the percent identity between two nucleotide sequences.
- a particularly preferred parameter set (and one that should be used unless otherwise stated) is the Blossum 62 scoring matrix with a gap penalty of 12, a gap extension penalty of 4, and a frameshift gap penalty of 5.
- nucleic acid sequences and protein sequences described herein can further be used as "query sequences" to perform searches against public databases, eg, to identify other family member sequences or related sequences.
- amino acid change and “amino acid modification” are used interchangeably to refer to additions, deletions, substitutions and other modifications of amino acids. Any combination of amino acid additions, deletions, substitutions and other modifications can be made, provided that the final polypeptide sequence possesses the desired properties.
- amino acid substitutions to the antibody result in decreased binding of the antibody to an Fc receptor.
- non-conservative amino acid substitutions ie the substitution of one amino acid with another amino acid having different structural and/or chemical properties, are particularly preferred.
- Amino acid substitutions include non-naturally occurring amino acids or naturally occurring amino acid derivatives of the twenty standard amino acids (e.g., 4-hydroxyproline, 3-methylhistidine, ornithine, homoserine, 5-hydroxy lysine) substitution.
- Amino acid changes can be made using genetic or chemical methods well known in the art. Genetic methods can include site-directed mutagenesis, PCR, gene synthesis, and the like. A method of changing amino acid side chain groups by methods other than genetic engineering, such as chemical modification, may be useful.
- Various names may be used herein to refer to the same amino acid change. For example, a substitution from proline to glycine at position 329 of the Fc domain may be denoted as 329G, G329, G329 , P329G Pro329Gly, or simply "PG".
- conservative sequence modification refers to an amino acid modification or change that does not significantly affect or alter the binding characteristics of an antibody or antibody fragment comprising an amino acid sequence.
- conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into antibodies or antibody fragments of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis.
- a conservative substitution is an amino acid substitution in which an amino acid residue is replaced by an amino acid residue with a similar side chain. Families of amino acid residues with similar side chains have been defined in the art.
- These families include those with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), ⁇ -side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenyl Alanine, tryptophan, histidine) amino acids.
- basic side chains e.g., lysine, arginine, histidine
- acidic side chains e.g., aspartic acid, glutamic acid
- autologous refers to any substance that is derived from the same individual into whom the substance is later reintroduced.
- allogeneic refers to any substance derived from a different animal of the same species as the individual into whom it is introduced. Two or more individuals are said to be allogeneic to each other when the genes at one or more loci are not identical. In some aspects, allogeneic material from individuals of the same species may be sufficiently dissimilar genetically to interact antigenically.
- xenogeneic refers to a graft derived from an animal of a different species.
- apheresis refers to an art-recognized extracorporeal method by which blood from a donor or patient is removed from a donor or patient and passed through a device that separates selected specific components and return the remainder to the donor or patient's circulation, for example, by retransfusion.
- single sample refers to a sample obtained using apheresis.
- immune effector cell refers to a cell that participates in an immune response, eg, participates in promoting an immune effector response.
- immune effector cells include T cells, eg, ⁇ / ⁇ T cells and ⁇ / ⁇ T cells, B cells, natural killer (NK) cells, natural killer T (NKT) cells, mast cells, and myeloid-derived phagocytes.
- immune effector function refers to, for example, the enhancement of immune effector cells or the function or response of immune attack target cells.
- immune effector function or response refers to T cell or NK cell properties that promote killing of target cells or inhibit growth or proliferation of target cells.
- primary stimulation and co-stimulation are examples of immune effector functions or responses.
- effector function refers to a specialized function of a cell.
- the effector function of T cells may be, for example, cytolytic activity or helper activity, including secretion of cytokines.
- T cell activation refers to one or more cellular responses of T lymphocytes, especially cytotoxic T lymphocytes, selected from the group consisting of proliferation, differentiation, secretion of cytokines, release of cytotoxic effector molecules, cytotoxic activity and activation Expression of markers.
- cytotoxic T lymphocytes selected from the group consisting of proliferation, differentiation, secretion of cytokines, release of cytotoxic effector molecules, cytotoxic activity and activation Expression of markers.
- the chimeric antigen receptors of the invention are capable of inducing T cell activation. Suitable assays for measuring T cell activation are described in the Examples and are known in the art.
- lentivirus refers to a genus of the family Retroviridae. Lentiviruses are unique among retroviruses in their ability to infect non-dividing cells; they can deliver significant amounts of genetic information to host cells, making them one of the most efficient methods of gene delivery vectors. HIV, SIV and FIV are examples of lentiviruses.
- lentiviral vector refers to a vector derived from at least a portion of a lentiviral genome, including inter alia self-inactivating lentiviral vectors as provided in Milone et al., Mol. Ther. 17(8):1453-1464 (2009).
- Other examples of lentiviral vectors that may be used clinically include, but are not limited to, the Lentiviral vector from Oxford BioMedica Gene delivery technology, LENTIMAX TM vector system from Lentigen, etc.
- Non-clinical types of lentiviral vectors are also available and known to those skilled in the art.
- BCMA-associated disease refers to any condition caused by, aggravated by, or otherwise associated with increased expression or activity of BCMA.
- mammals include, but are not limited to, domesticated animals (e.g., cattle, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rodents). mouse).
- domesticated animals e.g., cattle, sheep, cats, dogs, and horses
- primates e.g., humans and non-human primates such as monkeys
- rabbits e.g., mice and rodents.
- rodents e.g., mice and rodents.
- an individual or subject is a human.
- tumor and cancer are used interchangeably herein to encompass both solid and liquid tumors.
- cancer and “cancerous” refer to the physiological disorder of unregulated cell growth in mammals.
- tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and to all pre-cancerous and cancerous cells and tissues.
- cancer cancer
- cancer cancerous and cancerous cells and tissues.
- Tumor immune evasion refers to the process by which tumors evade immune recognition and elimination. Thus, as a therapeutic concept, tumor immunity is “cured” when such evasions are weakened, and the tumor is recognized and attacked by the immune system. Examples of tumor recognition include tumor binding, tumor shrinkage, and tumor clearance.
- half effective concentration refers to the concentration of drug, antibody or poison that induces 50% of the response between baseline and maximum after a specified exposure time.
- FACS fluorescence activated cell sorting
- Such instruments include the FACS Star Plus, FACScan, and FACSort instruments from Becton Dickinson (Foster City, CA), the Epics C from the Coulter Epics Division (Hialeah, FL), and the MoFlo from Cytomation (Colorado Springs, Colorado).
- pharmaceutically acceptable excipient refers to diluents, adjuvants (such as Freund's adjuvant (complete and incomplete)), excipients, buffers or stabilizers, etc., which are administered together with the active substance.
- treating means slowing, interrupting, arresting, alleviating, stopping, reducing, or reversing the progression or severity of an existing symptom, disorder, condition, or disease. Desirable therapeutic effects include, but are not limited to, prevention of disease onset or recurrence, alleviation of symptoms, reduction of any direct or indirect pathological consequences of disease, prevention of metastasis, reduction of the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
- antibody molecules of the invention are used to delay the development of a disease or to slow the progression of a disease.
- an effective amount refers to such an amount or dose of an antibody or composition of the present invention that, after administration to a patient in single or multiple doses, produces the desired effect in a patient in need of treatment or prevention.
- An effective amount can be readily determined by the attending physician, who is skilled in the art, by considering various factors such as: the species of mammal; body weight, age and general health; the particular disease involved; the extent or severity of the disease; the individual The patient's response; the specific antibody administered; the mode of administration; the bioavailability characteristics of the formulation administered; the chosen dosing regimen; and the use of any concomitant therapy.
- a “therapeutically effective amount” refers to an amount effective, at dosages required, and for periods of time required, to achieve the desired therapeutic result.
- a therapeutically effective amount of an antibody or antibody fragment or composition thereof can vary depending on factors such as the disease state, age, sex and weight of the individual and the ability of the antibody or antibody portion to elicit a desired response in the individual.
- a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody fragment or composition thereof are outweighed by the therapeutically beneficial effects.
- a “therapeutically effective amount” preferably inhibits a measurable parameter (e.g., tumor growth rate, tumor volume, etc.) by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 50%, relative to an untreated subject. 60% or 70% and still more preferably at least about 80% or 90%.
- Compounds can be evaluated for their ability to inhibit a measurable parameter (eg, cancer) in animal model systems predictive of efficacy in human tumors.
- non-fixed combination refers to non-fixed combination products or fixed combination products, including but not limited to kits and pharmaceutical compositions.
- non-fixed combination means that the active ingredients (e.g., (i) P329G CAR-T cells, and (ii) P329G mutant antibody against BCMA) are combined in separate entities at the same time, without specific time limit or in the same or different Time intervals, sequential administration to a subject, wherein such administration provides effective treatment in the subject.
- fixed combination means that the combination of the P329G mutant antibody against BCMA and the P329G CAR-T cells of the present invention is administered to the patient at the same time in the form of a specific single dose.
- non-fixed combination means that the combination of the 329G mutant antibody against BCMA of the present invention and the P329G CAR-T cells is administered to the patient simultaneously, concurrently or sequentially as separate entities, without specific dose and time limit, wherein such administration Therapeutically effective levels of the drug combinations of the invention in a patient are provided.
- the drug combination is a non-fixed combination.
- combination therapy refers to the administration of two or more components to treat a cancer as described in this disclosure.
- Such administration includes co-administration of the components in a substantially simultaneous manner.
- such administration includes co-administration or separate administration or sequential administration for each active ingredient in multiple or in separate containers (eg, capsules, powders and liquids). Powders and/or liquids can be reconstituted or diluted to the desired dosage before administration.
- administering further comprises using the P329G mutant antibody against BCMA of the invention and the P329G CAR-T cells of the invention at about the same time, or in a sequential manner at different times. In either case, the treatment regimen will provide for the beneficial effect of the drug combination in treating the disorders or conditions described herein.
- vector refers to a nucleic acid molecule capable of multiplying another nucleic acid to which it has been linked.
- the term includes vectors that are self-replicating nucleic acid structures as well as vectors that integrate into the genome of a host cell into which they have been introduced. Some vectors are capable of directing the expression of nucleic acids to which they are operably linked. Such vectors are referred to herein as "expression vectors.”
- host cell refers to a cell into which an exogenous polynucleotide has been introduced, including the progeny of such cells.
- Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom, regardless of the number of passages. Progeny may not be identical in nucleic acid content to the parental cell, but may contain mutations. Mutant progeny screened or selected for the same function or biological activity in originally transformed cells are included herein.
- a host cell is any type of cellular system that can be used to produce an antibody molecule of the invention, including eukaryotic cells, eg, mammalian cells, insect cells, yeast cells; and prokaryotic cells, eg, E. coli cells.
- Host cells include cultured cells as well as cells within transgenic animals, transgenic plants, or cultured plant or animal tissues.
- Subject/patient sample refers to a collection of cells, tissues or body fluids obtained from a patient or subject.
- the source of the tissue or cell sample can be solid tissue like from fresh, frozen and/or preserved organ or tissue samples or biopsy samples or puncture samples; blood or any blood components; body fluids such as cerebrospinal fluid, amniotic fluid (amniotic fluid ), peritoneal fluid (ascites), or interstitial fluid; cells from any time during pregnancy or development of a subject.
- Tissue samples may contain compounds that are not naturally intermingled with tissue in nature, such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, and the like.
- tumor samples herein include, but are not limited to, tumor biopsy, fine needle aspirate, bronchial lavage fluid, pleural fluid (pleural effusion), sputum, urine, surgical specimen, circulating tumor cells, serum, plasma, circulating Plasma proteins in ascites, primary cell cultures or cell lines derived from tumors or exhibiting tumor-like properties, and preserved tumor samples such as formalin-fixed, paraffin-embedded tumor samples, or frozen tumors sample.
- treating in reference to a disease means alleviating the disease (ie, slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof), preventing or delaying the onset or development or progression of the disease.
- CAR chimeric antigen receptor
- the present invention relates to chimeric antigen receptor polypeptides capable of specifically binding to mutant Fc domains of antibodies directed against BCMA molecules.
- the chimeric antigen receptor of the present invention comprises a humanized anti-P329G mutation scFv sequence, and the scFv sequence can specifically bind to the Fc domain of an antibody comprising a P329G mutation, but cannot specifically bind to the Fc domain of an unmutated parental antibody. domain.
- the Fc domain of an antibody comprising the P329G mutation has reduced binding to an Fc receptor (eg, an Fc gamma receptor) compared to the binding to an Fc receptor of an unmutated parental antibody Fc domain.
- the recombinant CAR construct of the present invention comprises a sequence encoding a CAR, wherein the CAR comprises a humanized anti-P329G mutation scFv sequence, and the scFv sequence specifically binds to the Fc domain of an antibody with a P329G mutation.
- the scFv sequence in the CAR construct of the present invention comprises the following sequence:
- CDR L light chain complementarity determining region 1 shown in the amino acid sequence RSSTGAVTTSNYAN (SEQ ID NO: 22), or a variant of said CDR L1 with no more than 2 amino acid changes or no more than 1 amino acid change;
- amino acid changes are additions, deletions or substitutions of amino acids.
- the scFv can be connected with a signal peptide sequence at the N-terminus, for example, the signal peptide sequence shown in SEQ ID NO: 11, and the scFv can be connected at the C-terminus with a sequence as provided in SEQ ID NO: 14 or SEQ ID NO: 18
- a hinge/spacer sequence, a transmembrane region as provided in SEQ ID NO: 15, a co-stimulatory signal domain as in SEQ ID NO: 16 and an intracellular stimulatory signal comprising SEQ ID NO: 17 or a variant thereof Domains for example, where the individual domains are adjacent to each other and in the same open reading frame to form a single fusion protein.
- the scFv domain comprises (i) a heavy chain variable region comprising or at least 90%, 91%, 92%, 93%, 94%, 95%, the sequence of SEQ ID NO: 12, A sequence of 96%, 97%, 98% or 99% identity, and (ii) a light chain variable region comprising or at least 90%, 91%, 92%, 93% of the sequence of SEQ ID NO: 13 , 94%, 95%, 96%, 97%, 98% or 99% identical sequences;
- the scFv domain comprises (i) the heavy chain variable region set forth in SEQ ID NO: 12 and (ii) the light chain variable region set forth in SEQ ID NO: 13.
- the scFv domain further comprises a (Gly4-Ser)n linker, wherein n is 1, 2, 3, 4, 5 or 6, preferably 3 or 4.
- the light and heavy chain variable regions of the scFv may, for example, be in any of the following orientations: light chain variable region-linker-heavy chain variable region or heavy chain variable region-linker-light chain variable region.
- an exemplary CAR construct of the invention comprises a signal peptide sequence, a humanized anti-P329G mutant scFv sequence, a hinge/spacer, a transmembrane domain, an intracellular co-stimulatory signaling domain, and an intracellular stimulatory domain. signaling domain.
- the present invention provides the amino acid sequence of the full-length CAR polypeptide as SEQ ID NO: 1, as shown in the sequence listing.
- the present invention provides a recombinant nucleic acid construct comprising a nucleic acid molecule encoding the CAR of the present invention, for example, it comprises encoding the amino acid sequence shown in SEQ ID NO: 1 or having at least the same amino acid sequence as SEQ ID NO: 1
- CAR constructs encoding the present invention can be obtained using recombinant methods well known in the art.
- the nucleic acid of interest may be produced synthetically rather than by genetic recombination methods.
- the present invention includes retroviral vector constructs and lentiviral vector constructs expressing a CAR that can be directly transduced into cells.
- the nucleic acid sequence of the CAR construct of the invention is cloned into a lentiviral vector to generate a full-length CAR construct in a single coding frame, and the EF1 ⁇ promoter is used for expression.
- CAR polypeptides of the present invention may also be modified so as to vary in amino acid sequence but not in desired activity.
- additional nucleotide substitutions that result in amino acid substitutions at "non-essential" amino acid residues can be made to the CAR polypeptide.
- a non-essential amino acid residue in a molecule can be replaced with another amino acid residue from the same side chain family.
- an amino acid stretch can be substituted for a structurally similar stretch that differs in the order and composition of side chain family members, for example, conservative substitutions can be made in which the amino acid residue Substitute with an amino acid residue with a similar side chain.
- Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, , glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta branched side chains (e.g., threonine, valine, iso leucine) and aromatic side chains (eg, tyrosine, phenylalanine, tryptophan, histidine).
- basic side chains e.g., lysine, arginine, histidine
- acidic side chains e.g., aspartic acid
- the present invention contemplates the generation of functionally equivalent CAR polypeptide molecules, e.g., the VH or VL of the humanized anti-P329G mutant scFv sequence contained in the CAR can be modified to obtain at least 90 %, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical VH and having at least 90%, 91%, 92%, A VL that is 93%, 94%, 95%, 96%, 97%, 98% or 99% identical.
- the transmembrane domain contained in the CAR of the present invention is an anchored transmembrane domain, which is a component of a polypeptide chain capable of being integrated in a cell membrane. Transmembrane domains can be fused to other extracellular and/or intracellular polypeptide domains, which will also be confined to the cell membrane. In chimeric antigen receptor (CAR) polypeptides of the invention, the transmembrane domain confers membrane attachment to the CAR polypeptide of the invention.
- the CAR polypeptide of the present invention comprises at least one transmembrane domain, which may be derived from a natural source or a recombinant source, comprising predominantly hydrophobic residues such as leucine and valine.
- the domain may be derived from the transmembrane domain of a membrane-bound or transmembrane protein such as CD28, CD8 (eg, CD8 ⁇ , CD8 ⁇ ).
- the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 15.
- the transmembrane domain in the CAR of the invention is linked to the extracellular region of the CAR (ie, the humanized anti-P329G mutant scFv sequence) via a hinge/spacer.
- the hinge can be the hinge region of CD8 ⁇ , the hinge region of CD28.
- the hinge or spacer sequence comprises the amino acid sequence of SEQ ID NO: 18.
- the glycine-serine doublet also provides a particularly suitable linker as hinge/spacer.
- the linker comprises the amino acid sequence of GGGGS (SEQ ID NO: 14).
- the cytoplasmic domain comprised in the CAR of the present invention comprises an intracellular signaling domain.
- the intracellular signaling domain is capable of activating at least one effector function of the immune cell into which the CAR of the present invention has been introduced.
- intracellular signaling domains useful in the CARs of the invention include those of the T cell receptor (TCR) and co-receptors that act cooperatively to initiate signal transduction upon binding of the extracellular domain to the Fc domain of an antibody with a P329G mutation.
- TCR T cell receptor
- the CAR of the present invention is also designed with a co-stimulatory signal domain (CSD) that can generate co-stimulatory signals.
- CSD co-stimulatory signal domain
- Activation of T cells is mediated by two distinct classes of cytoplasmic signaling sequences: those that initiate antigen-dependent primary activation through the TCR (primary intracellular signaling domain) and those that act in an antigen-independent manner to provide co-stimulatory signals Those sequences of (secondary cytoplasmic domain, eg, co-stimulatory domain).
- the CAR of the present invention comprises a primary intracellular signaling domain, e.g., the primary signaling domain of CD3 ⁇ , e.g., the CD3 ⁇ signaling domain shown in SEQ ID NO: 17.
- the intracellular signaling domain in the CAR of the present invention also includes a secondary signaling domain (ie, co-stimulatory signaling domain).
- a co-stimulatory signaling domain refers to the portion of the CAR comprising the intracellular domain of a co-stimulatory molecule.
- Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that are required for lymphocytes to respond effectively to antigens.
- co-stimulatory molecules include, but are not limited to, CD28, 4-1BB (CD137), which cause co-stimulation that enhances the proliferation, effector function, and survival of human CART cells in vitro and enhances the anti-inflammatory effect of human T cells in vivo. tumor activity.
- the intracellular signaling sequences of the CAR of the present invention can be connected to each other in a random order or in a specified order.
- short oligopeptide linkers or polypeptide linkers can form linkages between intracellular signaling sequences.
- a glycine-serine doublet can be used as a suitable linker.
- single amino acids, eg, alanine, glycine can be used as suitable linkers.
- the intracellular signaling domain of the CAR of the present invention is designed to include a co-stimulatory signaling domain of 4-1BB and a stimulating signaling domain of CD3 ⁇ .
- the invention provides nucleic acid molecules encoding the CAR constructs described herein.
- the nucleic acid molecule is provided as a DNA construct.
- the present invention also provides a vector inserted with the CAR construct of the present invention.
- Expression of a natural or synthetic nucleic acid encoding a CAR is achieved by operably linking the nucleic acid encoding the CAR polypeptide to a promoter and incorporating the construct into an expression vector.
- Vectors may be suitable for replication and integration in eukaryotes. Common cloning vectors contain transcriptional and translational terminators, initiation sequences and promoters for regulating the expression of the desired nucleic acid sequence.
- retroviruses provide a convenient platform for gene delivery systems.
- the selected gene can be inserted into a vector and packaged in retroviral particles using techniques known in the art.
- the recombinant virus can then be isolated and delivered to cells of the subject in vivo or ex vivo.
- retroviral systems are known in the art.
- lentiviral vectors are used.
- Retroviruses such as lentiviruses are suitable tools for long-term gene transfer because they allow long-term, stable integration of the transgene and its propagation in progeny cells.
- Lentiviral vectors have the added advantage over vectors derived from onco-retroviruses, such as murine leukemia virus, because they can transduce non-proliferative cells, such as hepatocytes. They also have the added advantage of low immunogenicity.
- a retroviral vector may also be, for example, a gamma retroviral vector.
- a gamma retroviral vector may, for example, comprise a promoter, a packaging signal ( ⁇ ), a primer binding site (PBS), one or more (e.g., two) long terminal repeats (LTRs), and a transgene of interest, e.g., encoding a CAR gene.
- Gamma retroviral vectors may lack viral structural genes such as gag, pol, and env.
- a promoter capable of expressing a CAR transgene in mammalian T cells is the EF1a promoter.
- the native EF1a promoter drives expression of the alpha subunit of the elongation factor-1 complex, which is responsible for the enzymatic delivery of aminoacyl tRNAs to the ribosome.
- the EF1a promoter has been used extensively in mammalian expression plasmids and has been shown to efficiently drive CAR expression from transgenes cloned into lentiviral vectors. See, eg, Milone et al., Mol. Ther. 17(8):1453-1464 (2009).
- CMV immediate early cytomegalovirus
- This promoter sequence is a constitutively strong promoter sequence capable of driving high-level expression of any polynucleotide sequence operatively linked thereto.
- other constitutive promoter sequences can also be used, including but not limited to Simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) Long terminal repeat (LTR) promoters, MoMuLV promoters, avian leukemia virus promoters, Epstein-Barr virus immediate early promoters, Rous sarcoma virus promoters, and human gene promoters such as but not limited to the actin promoter , myosin promoter, elongation factor-1 ⁇ promoter, hemoglobin promoter and creatine kinase promoter. Additionally, the present invention should not be limited to the use of constitutive promoters. Inducible promoters are also contemplated as part of
- the invention provides methods of expressing a CAR construct of the invention in mammalian immune effector cells (eg, mammalian T cells or mammalian NK cells) and immune effector cells produced thereby.
- mammalian immune effector cells eg, mammalian T cells or mammalian NK cells
- a source of cells eg, immune effector cells, eg, T cells or NK cells
- T cells can be obtained from a variety of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors.
- T cells can be obtained from blood components collected from a subject using any technique known to those of skill in the art, such as Ficoll (TM ) separation.
- the cells from the circulating blood of the individual are obtained by apheresis.
- Apheresis products generally contain lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and platelets.
- cells collected by apheresis can be washed to remove the plasma fraction and to place the cells in a suitable buffer or medium for subsequent processing steps.
- the cells are washed with phosphate buffered saline (PBS).
- PBS phosphate buffered saline
- T cell subsets such as CD3+, CD28+, CD4+, CD8+, CD45RA+ and CD45RO+ T cells can be further isolated by positive or negative selection techniques.
- anti-CD3/anti-CD28 conjugated beads such as M-450CD3/CD28T
- the period of time is between about 30 minutes and 36 hours or longer. Longer incubation times can be used to isolate T cells wherever small numbers of T cells are present, such as for isolating tumor infiltrating lymphocytes (TILs) from tumor tissue or from immunocompromised individuals.
- TILs tumor infiltrating lymphocytes
- Enrichment of T cell populations can be accomplished through the process of negative selection using a combination of antibodies directed against surface markers unique to the negatively selected cells.
- One method is the sorting and/or selection of cells by means of negative magnetic immunoadhesion or flow cytometry using the presence of cells on negatively selected cells Monoclonal antibody cocktail for surface markers.
- the immune effector cells can be allogeneic immune effector cells, eg, T cells or NK cells.
- the cell can be an allogeneic T cell, e.g., one lacking expression of a functional T cell receptor (TCR) and/or human leukocyte antigen (HLA) (e.g., HLA class I and/or HLA class II) T cells.
- TCR T cell receptor
- HLA human leukocyte antigen
- a T cell lacking a functional TCR can, for example, be engineered such that it does not express any functional TCR on its surface; engineered such that it does not express one or more subunits that make up a functional TCR (e.g. engineered so that it does not express or exhibit reduced expression of TCR ⁇ , TCR ⁇ , TCR ⁇ , TCR ⁇ , TCR ⁇ and/or TCR ⁇ ); or engineered so that it produces very few functional TCRs on its surface.
- a T cell as described herein can, for example, be engineered such that it does not express functional HLA on its surface.
- T cells described herein can be engineered such that cell surface expression of HLA (e.g., HLA class I and/or HLA class II) is downregulated.
- HLA e.g., HLA class I and/or HLA class II
- downregulation of HLA can be achieved by reducing or eliminating beta-2 microglobulin (B2M) expression.
- the T cells may lack a functional TCR and a functional HLA, e.g., HLA class I and/or HLA class II.
- the cells transduced with the nucleic acid encoding the CAR of the present invention are proliferated, for example, the cells are proliferated in culture for 2 hours to about 14 days.
- the immune effector cells expressing CAR obtained after in vitro proliferation can be tested for effector function as described in the Examples.
- the invention provides antibodies that bind BCMA with high target specificity and high affinity, comprising a heavy chain variable region and a light chain variable region, wherein:
- the heavy chain variable region comprises CDR H1 shown in the amino acid sequence SSSYYWT (SEQ ID NO: 25) according to Kabat numbering, or no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H1 CDR H2 shown in the amino acid sequence SISIAGSTYYNPSLKS (SEQ ID NO: 26), or a variant of no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H2; and the amino acid sequence DRGDQILDV (SEQ ID NO:27) shown in CDR H3, or the variant of no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H3;
- the light chain variable region comprises the amino acid sequence RASQSISRYLN (SEQ ID NO:28) shown in the CDR L1, or the CDR L1 variants with no more than 2 amino acid changes or no more than 1 amino acid change; CDR L2, Or a variant of no more than 2 amino acid changes or no more than 1 amino acid change of the CDR L2; and
- the heavy chain variable region comprises CDR H1 shown in the amino acid sequence NDVIS (SEQ ID NO: 31) according to Kabat numbering, or no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H1 CDR H2 shown in amino acid sequence VIIPIFGIANYAQKFQG (SEQ ID NO: 32), or a variant of no more than 2 amino acid changes or no more than 1 amino acid change of said CDR H2; and amino acid sequence GRGYYSSWLHDI (SEQ ID NO:33) shown in CDR H3, or the variant of no more than 2 amino acid changes or no more than 1 amino acid change of the CDR H3;
- the light chain variable region comprises the amino acid sequence QASQDITNYLN (SEQ CDR L1 shown in ID NO:34), or a variant of no more than 2 amino acid changes or no more than 1 amino acid change of said CDR L1; CDR L2 shown in the amino acid sequence DASNLET (SEQ ID NO:35), Or a variant of no more than 2
- amino acid changes are additions, deletions or conservative amino acid substitutions of amino acids.
- an antibody that binds a BCMA molecule of the invention binds mammalian BCMA, eg, human, cynomolgus monkey, mouse, rat, and rabbit BCMA.
- an antibody that binds a BCMA molecule of the invention has one or more of the following properties:
- ADCP antibody-dependent cellular cytotoxicity and/or antibody-dependent cellular phagocytosis
- an antibody of the invention that binds a BCMA molecule comprises a heavy chain variable region and a light chain variable region that specifically binds BCMA, wherein:
- the heavy chain variable region comprises the sequence of SEQ ID NO: 2 or is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical thereto sequence
- the light chain variable region comprises the sequence of SEQ ID NO: 3 or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% thereof % identity sequence
- the heavy chain variable region comprises the sequence of SEQ ID NO: 9 or is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical thereto sequence
- the light chain variable region comprises the sequence of SEQ ID NO: 10 or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% thereof % identity sequence
- amino acid change in the sequence of at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity is preferably amino acid substitution, more preferably amino acid conservative substitution , preferably, said amino acid changes do not occur in the CDR regions.
- an antibody of the invention that binds a BCMA molecule is an IgG1, IgG2, IgG3 or IgG4 antibody; preferably, it is an IgG1 or IgG4 antibody; more preferably, it is an IgG1 antibody, e.g., a human IgG1 antibody.
- the antibodies provided herein that bind to a BCMA molecule comprise a mutant Fc domain wherein the amino acid at position P329 is mutated to glycine (G) according to EU numbering, the same as the Fc ⁇ of the Fc domain of the parental antibody that is not mutated.
- a mutant Fc domain has reduced Fc ⁇ receptor binding compared to receptor binding; for example, the mutant Fc domain is a mutant Fc domain of an IgG1, IgG2, IgG3 or IgG4 antibody, preferably, the mutant Fc domain is A mutant Fc domain of an IgG1 or IgG4 antibody; more preferably, the mutant Fc domain is a mutant Fc domain of an IgG1 antibody, for example, the mutant Fc domain is a mutant Fc domain of a human IgG1 antibody.
- Antibodies that bind to BCMA molecules containing the P329G mutant Fc domain cannot exert antibody-dependent cellular cytotoxicity by binding to Fc ⁇ receptors, nor can they exert antibody-dependent cellular phagocytosis (ADCP).
- the invention provides a nucleic acid encoding any of the above antibodies or fragments thereof or any chain thereof that binds a BCMA molecule.
- a vector comprising said nucleic acid is provided.
- the vector is an expression vector.
- a host cell comprising said nucleic acid or said vector is provided.
- the host cell is eukaryotic.
- the host cell is selected from yeast cells, mammalian cells (eg, CHO cells or 293 cells), or other cells suitable for the production of antibodies or antigen-binding fragments thereof.
- the host cell is prokaryotic.
- a nucleic acid of the invention comprises a nucleic acid encoding an antibody of the invention that binds a BCMA molecule.
- one or more vectors comprising the nucleic acid are provided.
- the vector is an expression vector, such as a eukaryotic expression vector.
- Vectors include, but are not limited to, viruses, plasmids, cosmids, lambda phage, or yeast artificial chromosomes (YACs).
- the vector is a pcDNA3.4 expression vector.
- the expression vector can be transfected or introduced into a suitable host cell.
- Various techniques can be used to achieve this, for example, protoplast fusion, calcium phosphate precipitation, electroporation, retroviral transduction, viral transfection, biolistic, lipid-based transfection or other conventional techniques.
- protoplast fusion cells are grown in culture and screened for appropriate activity. Methods and conditions for culturing the produced transfected cells and for recovering the produced antibody molecules are known to those skilled in the art and can be based on this specification and methods known in the prior art, depending on the particular expression vector and Mammalian host cell alteration or optimization.
- cells that have stably incorporated DNA into their chromosomes can be selected by introducing one or more markers that allow selection of transfected host cells.
- a marker can, for example, confer prototrophy, biocidal resistance (e.g., antibiotics), or heavy metal (e.g., copper) resistance, etc. to an auxotrophic host.
- the selectable marker gene can be directly linked to the DNA sequence to be expressed or introduced into the same cell by co-transformation. Additional elements may also be required for optimal synthesis of mRNA. These elements can include splicing signals, as well as transcriptional promoters, enhancers and termination signals.
- a host cell comprising a polynucleotide of the invention.
- host cells comprising an expression vector of the invention are provided.
- the host cell is selected from yeast cells, mammalian cells, or other cells suitable for the production of antibodies.
- Suitable host cells include prokaryotic microorganisms such as E. coli.
- the host cells can also be eukaryotic microorganisms such as filamentous fungi or yeast, or various eukaryotic cells such as insect cells and the like. Vertebrate cells can also be used as hosts.
- mammalian cell lines adapted for growth in suspension can be used.
- Examples of useful mammalian host cell lines include SV40 transformed monkey kidney CV1 line (COS-7); human embryonic kidney line (HEK293 or 293F cells), 293 cells, baby hamster kidney cells (BHK), monkey kidney cells (CV1 ), African green monkey kidney cells (VERO-76), human cervical cancer cells (HELA), canine kidney cells (MDCK), Buffalo rat liver cells (BRL 3A), human lung cells (W138), human liver cells (HepG2), Chinese hamster ovary cells (CHO cells), CHO-S cells, NSO cells, myeloma cell lines such as Y0, NSO, P3X63 and Sp2/0, etc.
- the host cells are CHO cells or HEK293 cells.
- the present invention provides a method for preparing an antibody (including a P329G mutant antibody) that binds to a BCMA molecule, wherein said method comprises a nucleic acid suitable for expressing said antibody (including a P329G mutant antibody) that binds to a BCMA molecule
- the host cell comprising the nucleic acid encoding the antibody binding to BCMA molecule (including P329G mutant antibody) or the expression vector comprising said nucleic acid is cultivated under the conditions, and optionally the antibody binding to BCMA molecule (including P329G mutant antibody) is isolated ).
- the method further comprises recovering antibodies (including P329G mutant antibodies) that bind to BCMA molecules from the host cells (or host cell culture medium).
- Antibodies that bind to BCMA molecules of the present invention can be analyzed by known prior art techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography, size exclusion, etc. Purification by resistance chromatography, etc. The actual conditions used to purify a particular protein will also depend on such factors as net charge, hydrophobicity, hydrophilicity, and will be apparent to those skilled in the art.
- the purity of the BCMA molecule-binding antibodies of the invention, including P329G mutant antibodies can be determined by any of a variety of well-known analytical methods, including size exclusion chromatography, gel electrophoresis, high performance liquid chromatography Chromatography, etc.
- Antibodies provided herein that bind to BCMA molecules, including P329G mutant antibodies, can be identified, screened or characterized for their physical/chemical properties and/or biological activity by a variety of assays known in the art.
- the antigen-binding activity of the BCMA-binding antibody (including the P329G mutant antibody) of the present invention is tested, for example, by known methods such as FACS, ELISA or Western blotting. Binding to BCMA can be assayed using methods known in the art, exemplary methods are disclosed herein.
- the binding of BCMA molecule-binding antibodies of the invention, including P329G mutant antibodies, to cell surface BCMA is determined using FACS.
- the invention also provides assays for identifying biologically active antibodies that bind BCMA molecules, including P329G mutant antibodies.
- Biological activity may include, for example, ADCC action, CDC action, and the like.
- Cells for use in any of the above in vitro assays include cell lines that either naturally express BCMA or have been engineered to express BCMA.
- the modified BCMA-expressing cell line is a cell line that does not express BCMA under normal circumstances and expresses BCMA after the DNA encoding BCMA is transfected into cells.
- the molecular switch-regulated chimeric antigen receptor of the present invention is a regulatable CAR that can control the activity of CAR.
- the present invention uses the Pro329Gly (the 329th proline of the Fc segment of the antibody is mutated to glycine according to the EU numbering, abbreviated as P329G) mutant antibody as the safety switch in the CAR treatment of the present invention.
- P329G mutant antibody does not exist, the CAR activity of the present invention is turned off; when the P329G mutant antibody exists, the CAR activity of the present invention is turned on; thus, the opening and closing of the CAR molecule activity of the present invention is controlled by the P329G mutant antibody regulation.
- the present invention provides a pharmaceutical combination comprising (i) immune effector cells (e.g., T cells, NK cells) expressing the molecular switch-regulated CAR polypeptide of the present invention; and (ii) specifically binding to BCMA Molecular P329G mutant antibody.
- the immune effector cells are T cells expressing the molecular switch regulatory CAR polypeptide of the present invention prepared from autologous T cells or allogeneic T cells, for example, the immune effector cells are prepared from T cells isolated from human PBMC T cells expressing the molecular switch regulatory CAR polypeptide of the present invention.
- the P329G mutant antibody is ADI-38497 PG Ab and/or ADI-38484 PG Ab.
- the present invention provides a pharmaceutical combination comprising (i) a nucleic acid molecule encoding the molecular switch-regulated CAR polypeptide of the present invention or a vector comprising the nucleic acid component; and (ii) specifically binding to a BCMA molecule P329G mutant antibody.
- the pharmaceutical combinations of the present invention optionally further comprise pharmaceutically acceptable excipients of suitable formulations.
- suitable formulations for example, (ii) in the pharmaceutical combination can be formulated according to conventional methods (for example, Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Easton, U.S.A).
- the pharmaceutical combination of the present invention is used to treat BCMA-related diseases, such as cancers that express or overexpress BCMA, such as relapsed/refractory multiple myeloma (relapsed/refractory multiple myeloma, RRMM).
- BCMA-related diseases such as cancers that express or overexpress BCMA, such as relapsed/refractory multiple myeloma (relapsed/refractory multiple myeloma, RRMM).
- the present invention provides the aforementioned pharmaceutical combination of the present invention, which is used in the treatment of BCMA-related diseases in subjects, such as cancers expressing or overexpressing BCMA, such as relapsed/refractory multiple Relapsed/refractory multiple myeloma (RRMM).
- BCMA-related diseases such as cancers expressing or overexpressing BCMA, such as relapsed/refractory multiple Relapsed/refractory multiple myeloma (RRMM).
- RRMM relapsed/refractory multiple Relapsed/refractory multiple myeloma
- the pharmaceutical combination of the present invention is used to treat a cancer that expresses or overexpresses BCMA in a subject and is capable of reducing the severity of at least one symptom or indication of cancer or inhibiting the growth of cancer cells, said
- the cancer is, for example, relapsed/refractory multiple myeloma (RRMM).
- the invention provides methods of treating a BCMA-associated disease (e.g., a cancer that expresses or overexpresses BCMA, such as relapsed/refractory multiple myeloma) in a subject comprising administering The subject is administered a therapeutically effective amount of the pharmaceutical combination of the invention.
- a BCMA-associated disease e.g., a cancer that expresses or overexpresses BCMA, such as relapsed/refractory multiple myeloma
- the present invention provides the use of the aforementioned pharmaceutical combination of the present invention in the preparation of medicines for treating BCMA-related diseases (for example, cancers that express or overexpress BCMA, such as relapsed/refractory multiple myeloma) use.
- BCMA-related diseases for example, cancers that express or overexpress BCMA, such as relapsed/refractory multiple myeloma
- the pharmaceutical combinations of the invention may also be administered to individuals whose cancer has been treated with one or more prior therapies but has subsequently relapsed or metastasized, for example, the cancer is relapsed/refractory multiple myeloma (RRMM) .
- RRMM multiple myeloma
- immune effector cells for example, T cells, NK cells
- P329G specifically binding to BCMA molecules in the pharmaceutical combination of the present invention The mutated antibody is for parenteral, transdermal, intracavity, intraarterial, intravenous, intrathecal administration, or injected directly into tissue or tumor.
- the (ii) P329G mutant antibody specifically binding to BCMA molecule in the pharmaceutical combination of the present invention is activated in (i) immune effector cells (for example, T cells, NK cells) expressing the molecular switch-regulated CAR polypeptide of the present invention. cells) before, at the same time or after administration.
- the (i) immune effector cells expressing the molecular switch-regulated CAR polypeptide of the present invention in the pharmaceutical combination of the present invention are T cells expressing the CAR polypeptide of the present invention prepared from autologous T cells or allogeneic T cells.
- Cell; (ii) the P329G mutant antibody specifically binding to BCMA molecule in the pharmaceutical combination of the present invention is any antibody specifically binding to BCMA molecule, which contains the P329G mutation.
- the P329G mutant antibody is ADI-38497 PG Ab and/or ADI-38484 PG Ab.
- the present invention does not limit the component (i) in the drug combination of the present invention.
- the order in which component (i) and component (ii) are administered to the subject does not limit the timing between the administration of component (i) and component (ii) in the pharmaceutical combination of the present invention to the subject . Therefore, (i) and (ii) in the pharmaceutical combination of the present invention may be administered separately, simultaneously or sequentially.
- the administration of the two components can be separated by 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours or 72 hours, or by an interval that is readily apparent to those skilled in the art. Any suitable time difference determined.
- administering intravenously on the first day, administering (ii) on the second day, and then administering (ii) multiple times at a certain frequency, while monitoring the in vivo PK concentration of (i) and the desired therapeutic efficacy endpoint, to determine whether Multiple administration of (i); or administration of (ii) on the first day, intravenous administration of (i) on the second day, and then multiple administrations of (ii) at a certain frequency, while monitoring the in vivo PK concentration of (i) and the expected Therapeutic efficacy endpoint, to determine whether to administer (i) multiple times; or to administer (i) and (ii) at intervals of 1 hour, 2 hours, 4 hours, 6 hours, and 12 hours on the same day, and then administer (ii) multiple times at a certain frequency ), while determining whether to administer (i) multiple times by monitoring the in vivo PK concentration of (i) and the desired therapeutic efficacy endpoint.
- component (i) in the pharmaceutical combination of the present invention when component (i) in the pharmaceutical combination of the present invention is an immune effector cell (for example, T cell, NK cell) expressing the molecular switch-regulated CAR polypeptide of the present invention, it also includes The case where fraction (i) and component (ii) were pre-incubated together prior to administration to a subject. Thus, the two components may be preincubated for 1 minute, 5 minutes, 10 minutes, 15 minutes, 30 minutes, 45 minutes or 1 hour, or any suitable time readily determined by a person skilled in the art, prior to administration.
- an immune effector cell for example, T cell, NK cell
- fraction (i) and component (ii) were pre-incubated together prior to administration to a subject.
- the two components may be preincubated for 1 minute, 5 minutes, 10 minutes, 15 minutes, 30 minutes, 45 minutes or 1 hour, or any suitable time readily determined by a person skilled in the art, prior to administration.
- the pharmaceutical combination of the present invention can be administered to a subject at an appropriate dose.
- the dosage regimen will be determined by the attending physician and clinical factors. As is well known in the medical arts, the dose for any one patient depends on many factors, including the patient's weight, body surface area, age, the particular compound being administered, sex, time and route of administration, general health, and concomitant other medications administered.
- the component (i) in the pharmaceutical combination of the present invention is an immune effector cell (for example, T cell, NK cell) expressing the molecular switch-regulated CAR polypeptide of the present invention
- the component (i) 1 ⁇ 10 6 cells/kg body weight-10 ⁇ 10 6 cells/kg body weight, such as 1 ⁇ 10 6 cells/kg body weight, 2 ⁇ 10 6 cells/kg body weight, 3 ⁇ 10 6 cells/kg body weight Body weight, 5 ⁇ 10 6 cells/kg body weight, 7 ⁇ 10 6 cells/kg body weight, 9 ⁇ 10 6 cells/kg body weight, 10 ⁇ 10 6 cells/kg body weight in single or multiple doses and administer component (ii) at 0.1-10 mg/kg, preferably 0.1 mg/kg, 0.3 mg/kg, 0.5 mg/kg, 1 mg/kg, 3 mg/kg, 5 mg/kg,
- the dosage unit form of 7 mg/kg, 9 mg/kg, 10 mg/kg is preferably administered to the subject parenterally, more preferably intravenously.
- administration of a pharmaceutical combination of the invention to an individual with cancer results in complete disappearance of the tumor. In some embodiments, administration of a pharmaceutical combination of the invention to an individual with cancer results in at least an 85% or greater reduction in tumor cells or tumor size.
- Tumor reduction can be measured by any method known in the art, such as X-ray, positron emission tomography (PET), computed tomography (CT), magnetic resonance imaging (MRI), cytology, histology, or molecular genetics analyze.
- the drug combination of the present invention can reduce the "on-target/off tumor" toxicity of CAR-T cells.
- the present invention provides a kit comprising the pharmaceutical combination of the present invention, preferably the kit is in the form of a pharmaceutical dosage unit. Dosage units may thus be presented according to a dosing regimen or interval between drug administrations.
- kit of parts of the invention comprises in the same package:
- immune effector cells for example, T cells, NK cells
- nucleic acid molecules encoding the molecular switch-regulated CAR polypeptide of the present invention
- vectors comprising the nucleic acid, and any combination of them;
- Example 1 CAR gene synthesis, construction of virus expression vector, preparation of P329G CAR-T cells and detection of CAR expression
- the P329G CAR molecule (SEQ ID NO: 1), also known as HuR968B CAR, was constructed, consisting of a signal peptide (SP) shown in SEQ ID NO: 11, a specific single-chain antibody fragment (VH-linker-VL, VH-linker-VL, It has the VH shown in SEQ ID NO:12, the linker sequence shown in SEQ ID NO:41, the VL shown in SEQ ID NO:13), the G4S hinge region shown in SEQ ID NO:14, SEQ ID NO:15 Fusion of the CD8 transmembrane domain (CD8TM) shown, the 41BB costimulatory domain (41BB-CSD) shown in SEQ ID NO: 16, and the intracellular activation domain of the CD3 ⁇ molecule (CD3 ⁇ SSD) shown in SEQ ID NO: 17 made.
- SP signal peptide
- VH-linker-VL VH-linker-VL
- VH-linker-VL VH-linker-VL
- Blue21CAR (SEQ ID NO: 8) directly targeting BCMA was constructed and used as a control.
- Blue21CAR contains signal peptide shown in SEQ ID NO:11, anti-BCMA single-chain antibody (from 11D53 clone), hinge region of CD8 ⁇ molecule shown in SEQ ID NO:18 and SEQ ID NO:15 from N-terminal to C-terminal.
- the lentivirus pellet was resuspended in the medium to obtain the lentivirus concentrate,
- Pan T Cell Isolation Kit human
- T cell culture medium to resuspend the T cells to a certain concentration.
- Density and add TransAct (Miltenyi, 130-111-160) for activation.
- the lentivirus is the lentivirus encoding P329G CAR (SEQ ID NO: 1) or the control traditional CAR (SEQ ID NO: 8)) and blow the T cells evenly; the second day Remove the viral supernatant by centrifugation and resuspend the cells in fresh T cell medium.
- Figure 1A shows the expression of CAR in CD3 + cells, CD4 + , and CD8 + T cell subsets after the two CARs constructed in Example 1-1 were used to transduce T cells, and the results showed that in these transduced T cells
- the positive rate of CAR expression is about 18%-29%.
- GSK company BCMA antibody clone J6M0 light and heavy chain variable region sequence Obtained from the US9273141B2 patent, GSK company BCMA antibody clone J6M0 light and heavy chain variable region sequence, as a control antibody (GSK IgG).
- the light and heavy chain variable region sequences of GSK IgG, ADI-38497, and ADI-38484 antibodies were synthesized from the whole gene, and loaded into human IgG1 heavy chain constant region containing WT (SEQ ID NO: 4) or human IgG1 containing P329G point mutation On the pcDNA3.4 expression vector (purchased from Shanghai Boying) of heavy chain constant region (SEQ ID NO:5) and kappa light chain constant region (SEQ ID NO:6).
- the light and heavy chain expression vectors were co-transfected into HEK293 cells through PEI at a molar ratio of 2:3, and culture supernatants were collected after 5-7 days of culture.
- the antibody-containing supernatant medium was purified in one step through a Protein A column, and then dialyzed against PBS. The concentration was detected by reading the absorbance value at 280nm with a NanoDrop instrument, and the purity of the sample was detected by SDS-PAGE and SEC-HPLC. Obtained GSK WT antibody, GSK PG antibody; ADI-38497WT antibody, ADI-38497 PG antibody; and ADI-38484WT antibody, ADI-38484 PG antibody.
- Antibodies with BCMA antibody clone ADI-38497 heavy chain variable region (SEQ ID NO: 2) and light chain variable region (SEQ ID NO: 3) sequences are also referred to as ADI-38497 antibodies in this application, including ADI- The 38497 PG antibody and the ADI-38497WT antibody; antibodies with the heavy chain variable region (SEQ ID NO:9) and light chain variable region (SEQ ID NO:10) sequences of BCMA antibody clone ADI-38484 are also referred to in this application as It is ADI-38484 antibody, including ADI-38484 PG antibody and ADI-38484WT antibody.
- the specific method is as follows: After coupling anti-human Fc IgG (Ab97221, Abcam) to the surface of a CM5 chip (29149603, Cytiva), the ADI-38497 PG antibody was captured on the chip surface, and the BCMA antigen in the mobile phase was detected by detecting the antibody on the chip surface and the BCMA antigen in the mobile phase. Binding and dissociation between the obtained affinity and kinetic constants.
- the assay process used 10 ⁇ HBS-EP+(BR-1006-69, Cytiva) diluted 10 times as the experimental buffer. Each cycle in the affinity assay consists of capture of the ADI-38497 PG antibody, binding of one concentration of antigen, and regeneration of the chip.
- the antigen after gradient dilution (antigen concentration gradient is 1.25-40nM, 2-fold dilution) flows over the surface of the chip from low concentration to high concentration at a flow rate of 30 ⁇ l/min, the binding time is 180s, and an appropriate dissociation is set. Time (900s or 600s or 60s). Finally, the chip was regenerated using 10 mM glycine-HCl, pH 1.5 (BR-1003-54, Cytiva).
- Figure 2B shows representative affinity profiles of the ADI-38497 PG antibody to recombinant human, cynomolgus monkey, mouse, rat, and rabbit BCMA proteins measured by SPR.
- the results showed that the ADI-38497 PG antibody could bind to the above-mentioned BCMA proteins from different species, and the order of binding activity was human BCMA>monkey BCMA>mouse BCMA>rat BCMA>rabbit BCMA.
- CHO GS cells expressing BCMA antigens from different species were prepared. Specifically, BCMA genes derived from human, mouse, and cynomolgus monkeys were synthesized and cloned into lentiviral vectors, and then lentiviruses containing BCMA genes from different species were packaged, and CHO GS cells were infected with the lentiviruses.
- CHO GS cell lines expressing BCMA antigens from different species were obtained by flow cytometry sorting, namely hBCMA-CHO GS, mBCMA-CHO GS and cynoBCMA-CHO GS cells.
- the P329G antibody bound to the cells was detected by flow cytometry, and the APC channel MFI was analyzed, with the antibody concentration as the X axis and the APC channel MFI as the Y axis for drawing and calculating the binding EC50.
- Figure 2C shows the binding ability of different concentrations of P329G BCMA antibody to CHO-GS cells stably expressing human, cynomolgus monkey and mouse BCMA. It can be seen from Figure 2C that the ADI-38497 PG IgG antibody can bind to different species of BCMA expressed on the cell surface, while the BCMA antibody (Benchmark) derived from GSK has a high species-specificity for BCMA, and it does not recognize mouse BCMA. The results were consistent with the SPR detection results.
- Figure 2D shows the relationship between different concentrations of P329G BCMA antibody and positive multiple myeloma cell lines MM.1s, RPMI8226, U266, H929, L363 and AMO1 expressing BCMA
- MM.1s was purchased from Nanjing Kebai Biotechnology Co., Ltd., CBP60239
- RPMI8226 was purchased From Nanjing Kebai Biotechnology Co., Ltd., CBP60244
- U266 was purchased from Wuhan Punuosheng Life Technology Co., Ltd., CL-0510
- H929 was purchased from Nanjing Kebai Biotechnology Co., Ltd., CBP60243
- L363 was purchased from Nanjing Kebai Biotechnology Co., Ltd., CBP6024
- AMO1 was purchased from Nanjing Kebai Biotechnology Co., Ltd., CBP60242) binding activity, ADI-38497 PG antibody, ADI-38484 PG antibody can bind to positive tumor cells expressing BCMA in a concentration
- FIG. 3A shows a schematic diagram of the detection of the affinity of the specific single-chain antibody against the P329G mutation-rabbit Fc fusion protein and the ADI-38497 P329G mutation antibody by surface plasmon resonance (SPR).
- SPR surface plasmon resonance
- the specific method is as follows: After coupling the synthetic anti-PG scFv fusion protein (SEQ ID NO: 7) to the surface of C1 chip (BR100535, Cytiva), the binding and dissociation of the antibody on the chip surface and the ADI-38497 molecule in the mobile phase was detected. Obtain affinity and kinetic constants.
- the assay process used 10 ⁇ HBS-EP+(BR-1006-69, Cytiva) diluted 10 times as the experimental buffer. Each cycle of affinity detection includes binding of one concentration of antibody and regeneration of the chip.
- the serially diluted ADI-38497 molecules (concentration gradient 3.125nM-100nM, 2-fold dilution) flowed over the surface of the chip at a flow rate of 30 ⁇ l/min from low concentration to high concentration, the binding time was 180s, and the dissociation time was 180s. 300s. Finally, the chip was regenerated using 10mM Glycine-HCl, pH 1.5 (BR-1003-54, Cytiva).
- Figure 3B shows representative affinity profiles of ADI-38497 PG antibody and wild-type antibody binding to anti-PG scFv fusion protein by SPR. The results showed that only the ADI-38497 PG antibody could specifically bind to the anti-PG scFv as the extracellular domain of P329G CAR.
- FIG. 3C shows a schematic diagram of a method for measuring antibody affinity (Avidity) by surface plasmon resonance (SPR).
- the specific method is as follows: After coupling the anti-Fab IgG (I5260, Sigma) to the surface of the HLC chip (HLC30M, Xantec), capture the ADI-38497 molecule on the chip surface, and detect the fusion of the antibody on the chip surface with the anti-PG scFv in the mobile phase Affinities and kinetic constants are obtained for protein association and dissociation.
- the assay process used 10 ⁇ HBS-EP+(BR-1006-69, Cytiva) diluted 10 times as the experimental buffer. Each cycle of the avidity assay consists of capture of ADI-38497 molecules, binding of a concentration of anti-PG scFv, and regeneration of the chip.
- the anti-PG scFv after gradient dilution flows over the surface of the chip at a flow rate of 30 ⁇ l/min from low concentration to high concentration, the binding time is 180s, and the dissociation time is 300s .
- the chip was regenerated using 10 mM glycine-HCl, pH 1.5 (BR-1003-54, Cytiva).
- Figure 3D shows representative affinity profiles of ADI-38497 PG antibody and wild-type antibody with anti-PG scFv as P329G CAR extracellular domain measured by SPR. The results showed that only the ADI-38497 PG antibody could specifically bind to the P329G CAR.
- the ADI-38497 PG antibody and the wild-type ADI-38497WT antibody were prepared into 5-fold gradient diluted antibody solutions of different concentrations with FACS buffer, incubated with 1E5 CAR-positive cells at 4°C for 30 minutes, washed with FACS buffer, Incubate with Fc ⁇ fragment-specific APC goat anti-human IgG (Jackson ImmunoResearch, 109-136-098) for 30 minutes at 4°C.
- Flow cytometry was used to detect the antibody bound to the cells, and the APC channel MFI was analyzed, with the antibody concentration as the X-axis and the APC channel MFI as the Y-axis for plotting and calculating the binding EC50.
- Figure 3E shows the binding ability of ADI-38497WT antibody and ADI-38497 PG antibody to P329G CAR-T cells. The results showed that only the P329G mutant antibody showed binding to CAR, while the WT antibody did not, which was consistent with the SPR results.
- Table 8 summarizes the binding EC50 and EC90 values of ADI-38497 PG antibody and WT antibody to P329G CAR-T cells from different donors.
- PBMC cells Peripheral Blood Mononuclear Cells, peripheral blood mononuclear cells
- PBMC cells Peripheral Blood Mononuclear Cells, peripheral blood mononuclear cells
- the killing effect on the target cells is plotted and analyzed with the antibody concentration as the X-axis and the cell lysis ratio as the Y-axis.
- the cells were collected, washed twice with FACS buffer, and CD3, CD56, CD16 and CD107a antibodies were added, among which the CD107a antibody should be added in advance, and incubated with the cells at 37°C for 1 hour.
- the above cell-antibody mixture was stained at 4°C for 30 minutes, washed twice, resuspended in FACS buffer, and detected by flow cytometry.
- Figure 4A shows the ability of ADI-38497WT antibody and ADI-38497 PG antibody to mediate ADCC killing.
- CD3, CD56, CD16 and CD107a expression all showed that only the WT antibody mediated the ADCC cytotoxic killing effect on the positive H929 tumor cells expressing BCMA, while the P329G mutant antibody lacked the ability to induce the ADCC effect.
- ADCP reporter cell line Promega, G9871
- H929 cells in the logarithmic growth phase
- mix the ADCP reporter cells and H929 target cells according to the effect-to-target ratio of 2:1, 5:1, and mix with different concentrations of BCMA antibody, 37
- luciferase detection kit Promega, E2620
- Figure 4B shows the ability of ADI-38497WT antibody and ADI-38497 PG antibody to mediate ADCP killing.
- the results showed that when different effect-to-target ratios (2:1 or 5:1) were tested, only the ADI-38497WT antibody mediated the ADCP killing effect on BCMA-expressing positive H929 tumor cells, while the P329G mutant antibody lacked the effect on the expression of The ability of BCMA-positive H929 tumor cells to induce ADCP killing effect.
- H929 cells and L363 cells in the logarithmic growth phase were taken and spread in a certain number of well plates; a part of the H929 cells and L363 cells in the logarithmic growth phase were treated as target cells and treated with mitomycin C as a negative control. Then add different concentrations of ADI-38497 PG antibody to mix, continue to culture at 37°C for 48 hours, 72 hours and 120 hours respectively, use CellTiter-Glo (Promega, G9242) to detect the proportion of living cells, take the co-incubation time as the X axis, and the fluorescence The readings are plotted and analyzed on the Y-axis.
- Figure 4C shows the ability of ADI-38497 PG antibody to mediate target cell lysis. /ml), all showed that only the ADI-38497 PG antibody lacked the ability to induce target cell lysis.
- the CAR-T cells prepared by Donor 5 in Example 1 were revived and cultured at 37°C for overnight stability.
- centrifuge to collect cells wash twice with FACS buffer, resuspend and add goat anti-human IgG containing LIVE/DEAD Fixable Dead Cell Stain, Biotin-F(ab') Fragment (Jackson ImmunoResearch, 109- 066-006) FACS buffer, stained at 4°C for 30 minutes, washed twice, added CD4, CD8, CD25, CD69 and APC-Streptavidin antibody combination, the above cell antibody mixture was stained at 4°C for 30 minutes, washed twice, The cells were resuspended in FACS buffer
- Figure 5A shows that for H929 cells, only the ADI-38497 PG antibody containing the P329G mutation can specifically mediate the activation of CAR + -T, upregulate the expression levels of CD25 and CD69, and have no effect on CD4 + CAR + and CD8 + CAR + cells Differentially activated and exhibits a concentration gradient dependence of ADI-38497 PG antibody.
- FIG. 5B shows that different BCMA antibodies (ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, GSK PG IgG) induced the activation effect of L363 target cells on HuR968B CAR-T cells. It can be seen from Figure 5B that BCMA antibodies containing the P329G mutation (ADI-38497 PG antibody, ADI-38484 PG antibody, GSK PG IgG) can specifically mediate the activation of CAR+ T cells and significantly up-regulate the expression levels of CD25 and CD69. The expressions of CD25 and CD69 were also slightly up-regulated in the CAR-negative cell population, indicating a lower level of activation, but the degree of activation was not significant and could be ignored.
- BCMA antibodies containing the P329G mutation ADI-38497 PG antibody, ADI-38484 PG antibody, GSK PG IgG
- BCMA antibody containing the P329G mutation (ADI-38497 PG antibody, ADI- 38484 PG antibody, GSK PG IgG) can specifically mediate the activation of CAR + -T, significantly up-regulate the expression levels of CD25 and CD69, and have a gradient dependence of BCMA antibody with P329G mutation.
- the UNT cells and CAR-T cells prepared by Donor 4 in Example 1 were revived and cultured at 37°C for overnight stability. Dilute ADI-38497WT antibody or ADI-38497 PG antibody in PBS, add to 96-well plate and incubate overnight at 4°C to coat the plate. The CAR-T cells recovered after overnight culture were added to a 96-well plate coated with antibodies.
- CD3/CD28-coupled magnetic beads were directly added to the CAR-T cells (magnetic beads: cell ratio 3: 1); respectively incubated at 37°C for 72 hours and 120 hours, using Luminescent Cell Viability Assay (Promega, G7572) detects the luminescence value of cells.
- FIG. 5C shows the proliferation of HuR968B CAR-T cells stimulated by coated ADI-38497WT antibody or ADI-38497 PG antibody.
- HuR968B CAR-T cells proliferated under the stimulation of the coated ADI-38497 P329G antibody.
- ADI-38497 PG antibody made HuR968B CAR-T cells proliferate after 3 or 5 days of stimulation, respectively
- the stimulation with ADI-38497 PG antibody did not lead to significant proliferation of UNT cells under the stimulation of ADI-38497 PG antibody.
- Both HuR968B CAR-T cells and UNT cells proliferated significantly under the stimulation of CD3/CD28 antibody-coupled magnetic beads.
- the CAR-T cells prepared in Example 1 were revived and cultured at 37° C. overnight.
- the tumor target cells and CAR-T cells were mixed according to the E:T ratio of 2:1, and 5-fold or 10-fold serial dilutions of different concentrations of BCMA antibodies (ADI-38497PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody , or as Benchmark’s GSK PG IgG) solution to a total volume of 200 ⁇ L, incubated at 37°C for about 24 hours, centrifuged, and collected the supernatant.
- Cytokines were detected using BD TM Cytometric Bead Array (CBA) Human Th1/Th2 Cytokine Kit II ((BD, 551809)).
- Capture Beads in the kit, and distribute the plate at 25 ⁇ L/well.
- Figure 5D shows that the HuR968B CAR-T cells from donor 4 in Example 1 were co-cultured with H929 cells and RPMI8226 cells, after adding different concentrations of BCMA antibodies (ADI-38497 PG antibodies, ADI-38484 PG antibodies, ADI-38497WT antibodies, Or the release results of the effector cytokines secreted by CAR-T cells after GSK PG IgG as a positive control.
- BCMA antibodies ADI-38497 PG antibodies, ADI-38484 PG antibodies, ADI-38497WT antibodies
- FIG. 5E shows that HuR968B CAR-T cells from Donor 5 (purchased from ORiCELLS, Cat NO.: FPB004F-C, Lot NO.: PCH20210100004, Donor ID: Z0086) were co-cultured with different tumor cells, after adding different concentrations of BCMA antibody (ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or Benchmark GSK PG IgG) the release results of CAR-T cells secreting effector cytokines.
- BCMA antibody ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or Benchmark GSK PG IgG
- the CAR-T cells prepared in Example 1 were revived and cultured at 37° C. overnight.
- the tumor target cells and CAR-T cells were mixed according to the E:T ratio of 2:1, and different concentrations of BCMA antibodies (ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or Benchmark GSK PG IgG) solution to a total volume of 200 ⁇ L, cultured at 37°C for about 24 hours, centrifuged, and transferred the cell supernatant to a 96-well white-bottomed microplate.
- Use CytoTox 96 Non-Radioactive Cytotoxicity Assay (Promega, G1780) and microplate reader (Molecular Devices, SpectraMax i3x) to measure the LDH value in the supernatant, and calculate the killing efficiency.
- FIG. 5F shows that different BCMA antibodies (ADI-38497 PG antibody, ADI-38484 PG antibody, ADI-38497WT antibody, or Benchmark GSK PG IgG) induced HuR968B CAR-T cells to treat tumor cells with different BCMA expression levels (H929 ++ cells, Killing effect of RPMI8226 +++ cells, AMO1 + cells and L363 + cells).
- HuR968B cells were not activated and did not produce a killing effect on tumor cells.
- Figure 5G shows that neither the P329G BCMA antibody nor the WT BCMA antibody can mediate the killing effect of CAR-T on BCMA-negative target cells (such as BCMA-KO-H929 cells), while targeting BCMA-expressing H929 cells (H929) , WT BCMA antibody can not mediate the killing effect, only P329G BCMA antibody can induce CAR-T to produce a killing effect on tumor cells.
- BCMA-negative target cells such as BCMA-KO-H929 cells
- the CAR-T cells prepared by Donor 6 in Example 1 were revived and cultured at 37°C for overnight stability.
- the tumor target cells and CAR-T cells are mixed according to the E:T ratio of 2:1, and a certain concentration of PG BCMA antibody is added.
- BCMA antibody does not need to be added.
- CytoTox 96 Non-Radioactive Cytotoxicity Assay Promega, G1780
- a microplate reader Molecular Devices, SpectraMax i3x
- Figure 6 shows the effect of different concentrations of free BCMA protein on the killing effect of HuR968B CAR-T and Blue21CAR-T cells.
- PG BCMA antibody combined with soluble Combined with BCMA
- PG CAR-T can still exert normal killing effect. Therefore, the advantage of PG CAR-T cells is that their function is less affected by soluble ligands (such as soluble BCMA).
- BALB/c mice (age 4-6 weeks, body weight 15-17g, female) were divided into 3 groups, namely ADI-38497 PG antibody, 1mg/kg antibody group; ADI-38497 PG antibody, 10mg/kg antibody group; and ADI-38497 PG antibody, 200mg/kg antibody group, 9 mice in each group; the antibody was diluted to 0.1mg/mL, 1mg/mL and 20mg/mL with 1 ⁇ PBS, and the administration volume of each mouse was 10mL/ kg, that is, the antibody doses are 1mg/kg, 10mg/mL and 200mg/mL respectively; the administration method is intravenous injection, and the administration frequency is single.
- Coat the 96-well ELISA plate one day in advance Dilute the BCMA antigen to 1 ⁇ g/ml with coating solution (take a pack of carbonate (Thermo, 28382) powder, dissolve in 400 mL ultrapure water, dilute to 500 mL, and mix well to obtain the coating solution), 100 ⁇ L per well , seal the plate with film, and leave overnight at room temperature. Pour off the coating solution, pat dry on absorbent paper, then add 300 ⁇ L of washing solution to each well, shake and mix for 10 seconds, pat dry the washing solution, and repeat washing 3 times. Add blocking solution to each hole with a row gun, 200 ⁇ L, seal the plate with a sealing film, and incubate at room temperature for 2 hours. Then wash the plate once.
- coating solution take a pack of carbonate (Thermo, 28382) powder, dissolve in 400 mL ultrapure water, dilute to 500 mL, and mix well to obtain the coating solution
- 100 ⁇ L per well seal the plate with film,
- diluted standard curve graded with known concentration of BCMA antibody to prepare the standard curve (for example, use the known concentration of ADI-38497 PG antibody to prepare the standard curve), quality control samples and samples to be tested in 100 ⁇ L per well, at room temperature Incubate for 2 hours. Pour off the pre-coating solution, pat dry on absorbent paper, then add 300 ⁇ L of washing solution to each well, shake and mix for 10 seconds, pat dry the washing solution, and repeat washing 3 times. Repeat once. Goat anti-human IgG- Dilute Fc-HRP antibody (BETHYL) 1:100,000, add 100 ⁇ L to each well, incubate at room temperature in the dark for 1 hour. Then wash the plate once.
- BETHYL Goat anti-human IgG- Dilute Fc-HRP antibody
- TMB substrate to the 96-well ELISA plate, 100 ⁇ L per well, and incubate at room temperature in the dark. Light color development for 5 minutes. Add 50 ⁇ L ELISA stop solution to each well, shake for 10 seconds, and read OD450nm and OD620nm values within 30 minutes.
- FIGS 7A and 7B show the results of pharmacokinetic experiments of ADI-38497 PG antibody (hereinafter also referred to as PG Ab in vivo experiments in mice) in mice.
- ADI-38497 PG antibody 1mg/kg AUC0-inf, Cmax, CL, T1/2 were 2480 ⁇ g ⁇ h/mL, 30ug/ml, 0.40ml/kg/h, 145h;
- ADI -38497 PG antibody 10mg/kg: AUC0-inf, Cmax, CL, T1/2 are 24720 ⁇ g ⁇ h/mL, 187ug/ml, 0.32ml/kg/h, 219h; ADI-38497 PG antibody
- Example 8 The effect of PG CAR-T cells combined with different doses of PG antibody against BCMA high-expressing tumors in vivo
- mice Resuspend H929 cells with 1 ⁇ PBS to prepare a cell suspension with a cell concentration of 5 ⁇ 10 6 cells/mL.
- NOG mice (age 4-6 weeks, body weight 15-17g, female) were shaved on the right side of the back, subcutaneously injected with H929 cell suspension, the injection volume was 0.2mL/mouse, that is, the inoculation volume was 1 ⁇ 106 cells/mouse mouse.
- mice with tumor volumes ranging from 50.82 to 104.36 mm3 were divided into 7 groups, namely PBS vehicle group, PG Ab group, PG only CAR-T group, traditional CAR-T group, and PG Ab group.
- Ab+PG CAR-T 3mg/kg antibody group, PG Ab+PG CAR-T, 1mg/kg antibody group and PG Ab+PG CAR-T, 0.3mg/kg antibody group, 7 mice in each group.
- Antibodies with a concentration of 0.3mg/mL, 0.1mg/mL and 0.03mg/mL were respectively prepared. After the grouping was completed, the antibody was administered on the 7th day. The volume of administration per mouse was 10mL/kg, and the administration frequency was Once a week, the administration method is intraperitoneal injection.
- the CAR-T cells prepared by donor 4 were resuspended in 1 ⁇ PBS to prepare a CAR + cell suspension of 25 ⁇ 10 6 cells/mL, and 0.2 mL of the cell suspension was injected into the tail vein on the 7th day, and returned to Transfuse 5 ⁇ 10 6 CAR + cells/mouse.
- the body weight of the mice, the maximum long axis (L) and maximum width axis (W) of the tumor tissue were monitored twice a week.
- Figure 8A shows the therapeutic effect of different doses of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 high-expressing BCMA tumor cells.
- the results showed that in the tumor model with high expression of BCMA, the administration of PG CAR-T cells alone did not produce obvious anti-tumor effect, and only the administration of PG antibody produced a certain anti-tumor effect. Mice produced significant anti-tumor effects, and showed an antibody dose-dependent effect.
- TGI tumor growth inhibition
- Figure 8B shows the body weight changes of the mice in this experiment.
- the results showed that the body weight of mice treated with PG CAR-T cells combined with PG antibody remained stable after treatment, and the average body weight increased after treatment with PG antibody 0.3mg/kg, 1mg/kg and 3mg/kg 5.2%, 3.0%, 7.6%.
- the results showed that PG CAR-T cells combined with PG antibody therapy had a significant anti-tumor effect without obvious toxic side effects.
- Example 8-1 Take 30 ⁇ L of the mouse blood sample of Example 8-1, put it into a 96-well V-well plate, and mark it as the sample detection well; take 10 ⁇ L of the mouse blood sample, add it into the 96-well V-well plate, and mark it as the control well.
- Figure 8C shows the expansion of PG CAR-T cells in mice in the experiment of Example 8-1.
- the results showed that the in vivo expansion of PG CAR-T cells was dependent on the PG antibody, showing a certain dose-dependence of the antibody, and mice in the higher dose group had a higher level of PG CAR-T cell expansion.
- Blue21CAR-T cells used as a positive control showed similar expansion kinetics. They also began to expand after 1 week in the reinfused mice, and reached the peak level after 2 weeks (174769 cells/100 ⁇ L peripheral blood) , and remained at a very high level (131963 cells/100 ⁇ L peripheral blood) after 4 weeks.
- Example 9 The effect of PG CAR-T cells combined with different doses of PG antibody against BCMA low-expression tumors in vivo
- mice with tumor volumes ranging from 74.14 to 110.29 mm3 were divided into 7 groups, namely vehicle group, PG Ab group, PG CAR-T group, traditional CAR-T group, and PG Ab+ PG CAR-T, 3mg/kg antibody group, PG Ab+PG CAR-T, 1mg/kg antibody group, and PG Ab+PG CAR-T, 0.3mg/kg antibody group, with 7 mice in each group.
- Antibodies with a concentration of 0.3mg/mL, 0.1mg/mL and 0.03mg/mL were prepared respectively. After the grouping was completed, the antibody was administered on the 9th day.
- the volume of administration per mouse was 10mL/kg, and the administration frequency was Once a week, the administration method is intraperitoneal injection.
- the body weight of the mice, the maximum long axis (L) and maximum width axis (W) of the tumor tissue were monitored twice a week.
- Figure 9A shows the therapeutic effect of different doses of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human L363 low-expressing BCMA tumor cells.
- the results showed that in the tumor model with low expression of BCMA, only administration of PG CAR-T did not produce anti-tumor effect, while only administration of PG antibody had no obvious anti-tumor effect, with a TGI of 21%. Only the treated mice produced a significant anti-tumor effect, and showed an antibody dose-dependent effect.
- PG CAR-T cells combined with 0.3 mg/kg dose of PG antibody, PG CAR-T cells induced a significant anti-tumor effect, with a TGI of 87%.
- Figure 9B shows the body weight changes of the mice in this experiment.
- the results showed that the body weight of the mice treated with PG CAR-T cells combined with PG antibody maintained a steady increase after treatment, and the average body weight increased after the combination of PG antibody 0.3mg/kg, 1mg/kg, and 3mg/kg 18.2%, 10.5%, 8.5%.
- the results showed that PG CAR-T cells combined with PG antibody therapy had a significant anti-tumor effect and did not induce significant toxicity.
- Figure 9C shows the expansion of PG CAR-T cells in mice in the experiment of Example 9-1.
- the results showed that only PG CAR-T cells were administered, and they began to expand after 1 week of reinfusion into the mice, expanded to 919 cells/100 ⁇ L peripheral blood after 2 weeks, and rapidly decreased to 204 cells/100 ⁇ L peripheral blood at 3 weeks.
- peripheral blood 100 ⁇ L peripheral blood, 8049 cells/100 ⁇ L peripheral blood and 3347 cells/100 ⁇ L peripheral blood remained at high levels after 3 weeks, respectively 2475 cells/100 ⁇ L peripheral blood, 4121 cells/100 ⁇ L peripheral blood and 1969 cells Cells/100 ⁇ L of peripheral blood was much higher than that of the group without combined administration of antibodies during the same period.
- traditional Blue21CAR-T cells used as a positive control also expanded to a peak level (76836 cells/100 ⁇ L peripheral blood) after 2 weeks of reinfusion into mice, and maintained at a high level (36328 cells/100 ⁇ L peripheral blood) after 3 weeks. peripheral blood).
- mice Resuspend H929 cells with 1 ⁇ PBS to prepare a cell suspension with a cell concentration of 5 ⁇ 10 6 cells/mL.
- NOG mice (age 4-6 weeks, body weight 15-17g, female) were shaved on the right side of the back, subcutaneously injected with H929 cell suspension, the injection volume was 0.2mL/mouse, that is, the inoculation volume was 1 ⁇ 106 cells/mouse mouse.
- mice with tumor volumes ranging from 59.50 to 105.82 mm3 were divided into 7 groups, namely vehicle group, PG Ab group, PG CAR-T only group, and PG Ab+PG CAR-T group.
- mice per group 10 ⁇ 10 6 group, PG Ab+PG CAR-T, 1 ⁇ 10 6 cell group, PG Ab+PG CAR-T, 0.1 ⁇ 10 6 cell group and PG Ab+PG CAR-T, 0.01 ⁇ 10 6 cell groups, 7 mice per group.
- the CAR-T cells prepared from donor 4 were resuspended in 1 ⁇ PBS to prepare a CAR + cell suspension of 50 ⁇ 10 6 cells/mL, followed by 10-fold serial dilution, and then prepared into 5 ⁇ 10 6 , 0.5 ⁇ 10 6 and 0.05 ⁇ 10 6 cell suspensions/mL, inject 0.2 mL/mouse of the cell suspension into the tail vein on the 9th day.
- the body weight of the mice, the maximum long axis (L) and maximum width axis (W) of the tumor tissue were monitored twice a week.
- FIG. 10A shows the therapeutic effect of PG antibody combined with different doses of PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- the results showed that in the case of administering an extremely low dose of 0.01 ⁇ 10 6 CAR-T cells, CAR-T cells produced similar anti-tumor effects as those administered only with PG antibody, with TGIs of 49% and 50%, respectively.
- Increasing the dose of CAR-T cells to 0.1 ⁇ 10 6 , 1 ⁇ 10 6 , and 10 ⁇ 10 6 CAR-T cells significantly increased the anti-tumor effect induced by PG CAR-T cells, with TGIs of 91%, 104%, and 103, respectively.
- Administration of CAR-T cells alone did not show anti-tumor effects.
- Figure 10B shows the expansion of PG CAR-T cells in mice in the experiment of Example 10-1.
- the results showed that under the induction of PG antibody, CAR-T cells reinfused into the mice began to expand at 1 week, reached the peak level after 2 weeks, and remained at a high level after 3 weeks.
- the expansion of T cells in vivo depends on the dose of CAR-T cells.
- mice in the higher CAR-T dose group had higher levels of CAR-T cell expansion, 0.01 ⁇ 10 6 , 0.1 ⁇ 10 6 , 1 ⁇ 10 6 , 10
- the peak expansion levels of the ⁇ 10 6 CAR-T cell dose group were 6 cells/100 ⁇ L peripheral blood, 338 cells/100 ⁇ L peripheral blood, 3640 cells/100 ⁇ L peripheral blood, and 12895 cells/100 ⁇ L peripheral blood.
- Example 11 In vivo anti-tumor effect study of PG CAR-T cells combined with PG antibody at different administration frequencies
- H929 cells Resuspend H929 cells with 1 ⁇ PBS to prepare a cell suspension with a cell concentration of 5 ⁇ 10 6 cells/mL.
- NOG mice (age 4-6 weeks, body weight 15-17g, female) were shaved on the right side of the back, subcutaneously injected with H929 cell suspension, the injection volume was 0.2mL/mouse, that is, the inoculation volume was 1 ⁇ 106 cells/mouse mouse.
- mice with tumor volumes ranging from 51.25 to 94.97mm3 were divided into 7 groups, namely vehicle group, PG CAR-T only group, PG Ab+PG CAR-T, Q3/4 ⁇ 2 group, PG Ab+PG CAR-T, QW ⁇ 4 group, PG Ab+PG CAR-T, Q2W ⁇ 2 group, PG Ab+PG CAR-T, Q3W ⁇ 2 group and traditional CAR-T group, each Group of 7 mice.
- the PG Ab antibody with a concentration of 0.1 mg/mL was prepared, and after the grouping was completed, the antibody was administered, with a volume of 10 mL/kg administered to each mouse, and the administration method was intraperitoneal injection.
- the CAR-T cells prepared from donor 4 were resuspended in 1 ⁇ PBS to prepare a cell suspension of 10 ⁇ 106 CAR + cells/mL, and 0.2 mL of the cell suspension was injected into the tail vein.
- the body weight of the mice, the maximum long axis (L) and maximum width axis (W) of the tumor tissue were monitored twice a week.
- Fig. 11A shows the frequency of administration of the PG antibody in the experiment of Example 11-1.
- Q3-4D ⁇ 2 4 weeks as a cycle, PG antibody is administered twice in the first week, with an interval of 3-4 days;
- Q3W ⁇ 2 PG antibody is administered twice, and the dosing frequency is 3 weeks/time ;
- Figure 11B shows the therapeutic effect of different administration frequencies of PG antibody when combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- the results showed that when PG CAR-T cells were used in combination with different administration frequencies of PG antibody, PG CAR-T cells could be induced to produce significant anti-tumor effects, and the frequency of PG antibody administration was increased, and the anti-tumor effect induced by PG CAR-T cells was significant.
- the anti-tumor effect induced by CAR-T cells in the QW ⁇ 4 group was the most significant, showing the same anti-tumor effect as traditional Blue21 CAR-T cells.
- Figure 11C shows the expansion of PG CAR-T cells in mice in the experiment of Example 11-1.
- the results showed that when PG CAR-T cells were combined with PG antibody, the expansion kinetics of CAR-T cells were similar under the condition of different administration frequency of PG antibody. Expansion reached peak levels and declined to baseline levels after 3 weeks. In addition, the expansion of traditional Blue21CAR-T cells also reached the peak level 2 weeks after reinfusion into mice, and remained at a high level after 3 weeks.
- Example 12 Study on anti-systemic tumor effect of PG CAR-T cells combined with different doses of PG antibody in vivo
- H929-luc cells were prepared. Specifically, H929 cells (purchased from Nanjing Kebai Biotechnology Co., Ltd.) were used to package the lentivirus containing the GFP-luciferase gene, and the H929 cells were infected with the obtained lentivirus, and then sorted by flow cytometry to obtain H929-luc cell line expressing both GFP and luciferase.
- H929-luc cells were resuspend with 1 ⁇ PBS to prepare a cell suspension with a cell concentration of 25 ⁇ 10 6 cells/mL.
- NOG mice (age 4-6 weeks, body weight 15-17g, female) were injected with H929-luc cell suspension through the tail vein, with an injection volume of 0.2 mL/mouse.
- the substrate D-Luciferin (15mg/mL) was injected intraperitoneally, with an injection volume of 10mL/kg/mouse, and was analyzed by IVIS spectrum imaging 10 minutes after substrate injection.
- mice with fluorescence signals between 1.17 ⁇ 10 7 and 1.43 ⁇ 10 8 photons/sec into 7 groups, namely vehicle group, PG Ab, 0.3mg/kg group, PG Ab, 3mg/kg group, PG CAR-T group, PG Ab+PG CAR-T, 0.3mg/kg+2 ⁇ 10 6 group, PG Ab+PG CAR-T, 3mg/kg+2 ⁇ 10 6 group and Blue21CAR-T group, 6-7 animals in each group mice.
- Antibodies with a concentration of 0.03 mg/mL and 0.3 mg/mL were respectively prepared. After the grouping was completed, antibody administration was started on the 14th day.
- the volume of administration was 10 mL/kg for each mouse, and the administration frequency was once a week.
- the way of administration is intraperitoneal injection.
- the CAR-T cells prepared by donor 4 were resuspended in 1 ⁇ PBS to prepare a CAR + cell suspension of 10 ⁇ 10 6 cells/mL, and 0.2 mL of the cell suspension was injected into the tail vein on the seventh day.
- Figure 12A shows the fluorescence images of the therapeutic effects of different doses of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated with human H929-luc tumor cells in the tail vein.
- PG antibody combined with PG CAR-T cells also produced a significant anti-tumor effect.
- the fluorescence distribution and intensity of mice in the PG antibody 0.3 mg/kg combined with PG CAR-T cell group Compared with the control group, it was significantly reduced.
- mice in the PG antibody 3mg/kg combined with PG CAR-T cell group had no obvious fluorescence signal, while the traditional Blue21CAR-T treatment group still had a large amount of fluorescence distribution, showing that the PG CAR-T cell group was higher than the Blue21CAR-T cell group.
- Figure 12B shows the therapeutic effect of different doses of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated with human H929-luc tumor cells in the tail vein.
- PG antibody was administered at 0.3 mg/kg
- PG CAR-T cells produced a certain anti-tumor effect
- increasing the dose of PG antibody to 3 mg/kg the anti-tumor effect induced by PG CAR-T cells increased significantly, and the anti-tumor effect was significantly increased.
- the tumor effect was maintained longer, and the PG antibody was stopped, and the tumor growth began to recur in about 4 weeks, while the traditional Blue21CAR-T tumor relapsed faster when given the same cell dose, showing that PG CAR-T cells were more durable than traditional Blue21CAR-T antitumor effect. Only PG CAR-T cell group and PG antibody group did not show obvious anti-tumor effect.
- Figure 12C shows the change in body weight of the mice in the above experiment. The results showed that the weight of the mice in each treatment group increased steadily during the treatment period, suggesting that different doses of PG antibody combined with PG CAR-T cells did not induce significant toxicity in the hematological tumor model.
- H929 cells Resuspend H929 cells with 1 ⁇ PBS to prepare a cell suspension with a cell concentration of 5 ⁇ 10 6 cells/mL.
- NOG mice (age 4-6 weeks, body weight 15-17g, female) were shaved on the right back, and injected subcutaneously with 5 ⁇ 10 6 cells/mL of H929 cell suspension, with an injection volume of 0.2 mL/mouse.
- mice with tumor volumes ranging from 38.49 to 104.77mm3 were divided into 8 groups, as shown in Table 11, which were the non-tumor-bearing vehicle group, PG Ab+PG CAR-T, 10mg /kg+10 ⁇ 10 6 group; and tumor-bearing vehicle group, PG CAR-T group, PG Ab group, PG Ab+PG CAR-T, 10mg/kg+10 ⁇ 10 6 group, PG Ab+PG CAR -T, 3mg/kg+10 ⁇ 10 6 group and PG Ab+PG CAR-T, 3mg/kg+1 ⁇ 10 6 group, 24 mice in each group.
- Antibodies with a concentration of 1 mg/mL and 0.3 mg/mL were respectively prepared. After the grouping was completed, the antibody was administered, with a volume of 10 mL/kg administered to each mouse. The way of medicine is intraperitoneal injection.
- the CAR-T cells prepared by donor 4 were resuspended with 1 ⁇ PBS, and CAR + cells were prepared at 50 ⁇ 106 /mL and 5 ⁇ 106 /mL cell suspensions, respectively. Inject 0.2 mL of cell suspension into the tail vein.
- the body weight of the mice, the maximum long axis (L) and maximum width axis (W) of the tumor tissue were monitored twice a week. Peripheral blood was collected before the first antibody administration, after CAR-T reinfusion, before the third antibody administration, and at the end of the experiment. Four mice in each group were used for hematology and blood biochemical tests.
- Figure 13A shows the therapeutic effect of PG antibody combined with PG CAR-T cells in immunodeficient tumor-bearing mice inoculated subcutaneously with human H929 tumor cells.
- the results showed that the PG antibody combined with PG CAR-T cell treatment group produced significant anti-tumor effects, while the anti-tumor effect of the mice in the PG antibody-only treatment group was weak, and PG CAR-T alone did not produce anti-tumor effects.
- Figure 13B shows the change in body weight of the mice in this experiment.
- the results showed that the weight of the mice in the tumor-bearing and non-tumor-bearing treatment groups increased steadily during the treatment period, and there was no significant change in body weight compared with the control mice, suggesting that different doses of PG antibody combined with different doses of PG CAR-T cells did not induce obvious toxic reactions.
- Figure 13C and Figure 13D show the results of the mouse hematology and blood biochemical tests in the above experiments. The results showed that during the treatment period, the hematology and blood biochemical indicators of the mice in the tumor-bearing and non-tumor-bearing treatment groups had no significant changes compared with the control mice, indicating that the PG antibody combined with PG CAR-T cell therapy did not produce toxic reactions.
- Embodiment 14 clinical research
- the active components of the test drug described in this example are: P329G BCMA antibody (ADI-38497 PG antibody) and P329G CAR-T cells (the patient's own PBMC cells are transformed to obtain CAR-T cells).
- P329G BCMA antibody is 20.0mg/ml P329G BCMA antibody, 0.76mg/ml histidine, 1.08mg/ml histidine hydrochloride, 50.00mg/ml sorbitol, 0.20mg/ml polysorbate 80, pH 6.0 ;Specification of P329G BCMA antibody: 60mg (3mL)/bottle.
- Each bag contains 90-140 ⁇ 10 6 anti-P329G CAR-T positive cells dissolved in 7.5% DMSO cryoprotectant.
- P329G BCMA antibody The manufacturer of P329G BCMA antibody is Innovent Biopharmaceutical (Suzhou) Co., Ltd.; the manufacturer of P329G CAR-T cells is Innovent Cell Pharmaceutical (Suzhou) Co., Ltd.
- the proportion of monoclonal plasma cells detected by bone marrow cytology, bone marrow biopsy tissue or flow cytometry is ⁇ 5%;
- serum immunoglobulin free light chain ⁇ 10 mg/dL and serum immunoglobulin ⁇ / ⁇ free light chain ratio is abnormal.
- the ECOG score is 0 or 1.
- ANC absolute neutrophil count
- ALC absolute lymphocyte count
- platelets ⁇ 50 ⁇ 109/L
- Hemoglobin ⁇ 60g/L
- Renal function serum creatinine ⁇ 2.5 ⁇ ULN; or creatinine clearance (CrCl) ⁇ 40ml/min calculated according to the Cockcroft-Gault formula;
- Coagulation function fibrinogen ⁇ 1.0g/L; activated partial thromboplastin time, activated partial thromboplastin time ⁇ 1.5 ⁇ ULN, prothrombin time ⁇ 1.5 ⁇ ULN;
- left ventricular ejection fraction left ventricular ejection fraction (left ventricular ejection fraction, LVEF) ⁇ 50%.
- Subjects must sign the ICF, indicating that they understand the purpose and procedures of this study and are willing to participate in the study. Informed consent must be obtained before starting any study-related examination or procedure that is not part of the standard treatment of the subject's disease.
- This study is a clinical study to evaluate the safety, tolerability, pharmacokinetics and preliminary efficacy of infusion of "P329G BCMA and P329G CAR-T cells" in the treatment of RRMM subjects.
- the enrolled subjects will receive peripheral blood mononuclear cell (PBMC) apheresis (-28 ⁇ -21 days). Afterwards, the subject's own T cells were used to prepare P329G CAR-T cell preparations (hereinafter referred to as PG CAR-T cells). After successful preparation of PG CAR-T cells, the subject will first receive a preconditioning regimen of cyclophosphamide and fludarabine (-5 to -3 days), and then receive a dose of P329G 1 day after the rest assessment after pretreatment.
- PBMC peripheral blood mononuclear cell
- BCMA antibody (hereinafter referred to as ADI-38497 PG antibody) infusion, PG CAR-T cell infusion on the next day (0 day), and then from the PG CAR-T cell infusion day, periodic infusion of ADI every 21 days -38497 PG antibody, infused for up to 24 months, until disease progression, unacceptable toxicity, subject's withdrawal of informed consent, or other reasons for stopping study treatment (whichever occurs first).
- ADI-38497 PG antibody dosage has 3 dosage groups: 0.3mg/kg, 1mg/kg, 3mg/kg.
- the dose of PG CAR-T cells is 2*10 6 cells/kg.
- the DLT observation window is 21 days after the first dose of ADI-38497 PG antibody administration, and the DLT observation period includes 1 administration of PG CAR-T cells.
- Each dose group first enrolls 3 subjects, if no DLT is observed within the DLT observation window for the first 3 subjects, the administration of the next dose group can be started; if ⁇ 2 of the 3 subjects have DLT , the dose escalation will be terminated; if one of the 3 subjects who entered the group first at a certain dose has a DLT, 3 more subjects should be added to the same dose group (at this time, there are 6 subjects in this dose group in total). If no DLT occurred in the 3 supplementary subjects, the administration of the next dose group could be started. If ⁇ 1 of the 3 supplementary subjects had a DLT, the dose escalation was not allowed.
- the maximum tolerated dose MTD
- ⁇ 2 of the first 3 subjects in the initial antibody dose of 0.3 mg/kg group have DLT, they will return to a lower antibody dose of 0.1 mg/kg for dose exploration; If there is 1 case of DLT among the subjects, this dose group needs to add 3 subjects (at this time, there are 6 subjects in this dose group in total), if no DLT occurs in the 3 supplementary subjects, then the next trial can be started.
- Dosage group administration if ⁇ 1 case of DLT occurs in the 3 supplemented subjects, then the lower antibody dose of 0.1mg/kg will be returned for dose exploration.
- the investigator can choose the dose that may have a higher benefit/risk ratio to continue the study after discussing with the sponsor. For example, if the overall safety of the above-mentioned trials is good, it is predicted that further increasing the dose of PG CAR-T cells may increase the benefit of the subjects, and continue to explore the safety of the dose of 5*10 6 /kg PG CAR-T cells.
- the investigator will review the safety data, combined with the PK data, to decide whether it is necessary to explore the next dose group and adjust the dose escalation scheme if necessary.
- the first 3 subjects will adopt a staggered enrollment strategy. There must be an observation period of at least 2 weeks between the 1st and 2nd subjects, and the 2nd and 3rd subjects.
- the current dose group will continue to receive the treatment until the disease progresses, intolerable toxicity, the subject withdraws informed consent or other conditions that require stopping Reason for study treatment (whichever occurs first), with a maximum treatment period of 24 months.
- P329G BCMA antibody In the presence of a sufficient amount of P329G BCMA antibody, a very low dose of P329G CAR-T cells can induce a complete anti-tumor effect, and the effect is at least equivalent to that of The dose of traditional CAR-T cells was equivalent; the activity of P329G CAR-T cells was regulated by P329G BCMA antibody and showed a dose-dependent antibody dose, and different doses of P329G BCMA antibody induced different anti-tumor effects.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Immunology (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Zoology (AREA)
- Biophysics (AREA)
- Cell Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biomedical Technology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Hematology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Virology (AREA)
- Developmental Biology & Embryology (AREA)
- Oncology (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Mycology (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
PBMC细胞编号 | 目录号 | 批次号 | 供者ID号 |
供者3 PBMC | FPB004F-C | PCH2020110004 | Z0052 |
供者4 PBMC | FPB005F-C | PCH20201200002 | Z0066 |
供者5 PBMC | FPB004F-C | PCH20210100004 | Z0086 |
供者6 PBMC | FPB004F-C | PCH20201200043 | Z0084 |
供者7 PBMC | FPB004F-C | PCH20210100012 | Z0091 |
抗体 | EC50(nM) |
ADI-38497 PG | 0.3502至0.7433 |
ADI-38497 WT | 非常宽 |
组别 | 细胞系 | 处理 | 浓度 |
1 | 无肿瘤负荷 | 载剂 | N/A |
2 | 无肿瘤负荷 | PG Ab+PG CAR-T | 10mg/kg(QW×3)+10x 10 6个细胞/小鼠(SD) |
3 | H929 | 载剂 | N/A |
4 | H929 | PG Ab | 10mg/kg(QW×3) |
5 | H929 | PG CAR-T | 10x 10 6个细胞/小鼠(SD) |
6 | H929 | PG Ab+PG CAR-T | 10mg/kg(QW×3)+10x 10 6个细胞/小鼠(SD) |
7 | H929 | PG Ab+PG CAR-T | 3mg/kg(QW×3)+10x 10 6个细胞/小鼠(SD) |
8 | H929 | PG Ab+PG CAR-T | 3mg/kg(QW×3)+1x 10 6个细胞/小鼠(SD) |
Claims (15)
- 药物组合,其包含(i)第一组分,其选自表达分子开关调控型CAR多肽的免疫效应细胞(例如,T细胞、NK细胞)、编码所述CAR多肽的核酸分子、包含所述核酸分子的载体、和它们的任意组合;和(ii)第二组分,其是包含P329G突变的特异性结合BCMA分子的抗体或抗原结合片段(也称为P329G突变抗体),例如,所述P329G突变抗体包含突变Fc结构域,其中根据EU编号的P329位置处的氨基酸突变为甘氨酸(G),与未突变的亲本抗体Fc结构域的Fcγ受体结合相比,突变Fc结构域的Fcγ受体结合降低;以及任选地可药用辅料;其中,所述分子开关调控型CAR多肽包含(1)人源化抗P329G突变scFv序列,其中所述scFv序列包含能够特异性结合包含P329G突变的抗体Fc结构域,但不能特异性结合未突变的亲本抗体Fc结构域的如下序列:(i)重链可变区,其包含根据Kabat编号的(a)氨基酸序列RYWMN(SEQ ID NO:19)所示的重链互补决定区CDR H1、或所述CDR H1的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;(b)氨基酸序列EITPDSSTINYAPSLKG(SEQ ID NO:20)所示的CDR H2、或所述CDR H2的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;和(c)氨基酸序列PYDYGAWFAS(SEQ ID NO:21)所示的CDR H3、或所述CDR H3的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;和(ii)轻链可变区,其包含根据Kabat编号的(d)氨基酸序列RSSTGAVTTSNYAN(SEQ ID NO:22)所示的轻链互补决定区(CDR L)1、或所述CDR L1的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;(e)氨基酸序列GTNKRAP(SEQ ID NO:23)所示的CDR L2、或所述CDR L2的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;和(f)氨基酸序列ALWYSNHWV(SEQ ID NO:24)所示的CDR L3、或所述CDR L3的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;其中所述氨基酸变化是氨基酸的添加、缺失或取代;(2)铰链区/间隔区,其选自(i)(G 4S) n、(SG 4) n或G 4(SG 4) n肽接头,其中“n”是1至10的整数,例如1至4的整数;例如SEQ ID NO:14所示的序列;(ii)CD8α铰链区或其具有1-5个氨基酸修饰的变体,例如,SEQ ID NO:18所示的序列或其具有1-2个氨基酸修饰的变体;(3)跨膜区(TM),其选自CD8跨膜结构域或其具有1-5个氨基酸修饰的变体,例如,SEQ ID NO:15所示的序列或其具有1-2个氨基酸修饰的变体;(4)共刺激信号结构域(CSD),其选自4-1BB共刺激结构域或其具有1-5个氨基酸修饰的 变体,例如,SEQ ID NO:16所示的序列或其具有1-2个氨基酸修饰的变体;(5)刺激信号结构域(SSD),为CD3ζ信号传导结构域或其具有1-10个氨基酸修饰的变体,例如,SEQ ID NO:17所示的序列或其具有1-10个、1-5个氨基酸修饰的变体;优选地,所述CAR多肽包含(1)人源化抗P329G突变scFv序列,其中所述scFv序列包含能够特异性结合包含P329G突变的抗体Fc结构域,但不能特异性结合未突变的亲本抗体Fc结构域的如下序列:(i)重链可变区,其包含根据Kabat编号的(a)氨基酸序列RYWMN(SEQ ID NO:19)所示的CDR H1;(b)氨基酸序列EITPDSSTINYAPSLKG(SEQ ID NO:20)所示的CDR H2;和(c)氨基酸序列PYDYGAWFAS(SEQ ID NO:21)所示的CDR H3;和(ii)轻链可变区,其包含根据Kabat编号的(d)氨基酸序列RSSTGAVTTSNYAN(SEQ ID NO:22)所示的CDR L1;(e)氨基酸序列GTNKRAP(SEQ ID NO:23)所示的CDR L2;和(f)氨基酸序列ALWYSNHWV(SEQ ID NO:24)所示的CDR L3;例如,(i)重链可变区,其包含SEQ ID NO:12的序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列,和(ii)轻链可变区,其包含SEQ ID NO:13的序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列;例如,(i)重链可变区,其包含SEQ ID NO:12的序列,和(ii)轻链可变区,其包含SEQ ID NO:13的序列;(2)铰链区/间隔区,其选自(i)(G 4S) n、(SG 4) n或G 4(SG 4) n肽接头,其中“n”是1至4的整数,例如SEQ ID NO:14所示的序列;(ii)SEQ ID NO:18所示的CD8α铰链区序列或其具有1个氨基酸修饰的变体;(3)跨膜区(TM),其选自SEQ ID NO:15所示的CD8跨膜结构域或其具有1个氨基酸修饰的变体;(4)共刺激信号结构域(CSD),其选自SEQ ID NO:16所示的4-1BB共刺激结构域或其具有1个氨基酸修饰的变体;(5)刺激信号结构域(SSD),其选自SEQ ID NO:17所示的CD3ζ信号传导结构域或其具有1个氨基酸修饰的变体;其中所述氨基酸修饰是氨基酸的添加、缺失或取代。
- 根据权利要求1所述的药物组合,其中所述包含P329G突变的特异性结合BCMA分子的抗体或抗原结合片段包含重链可变区和轻链可变区,其中:(a)所述重链可变区包含根据Kabat编号的氨基酸序列SSSYYWT(SEQ ID NO:25)所示的 CDR H1、或所述CDR H1的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;氨基酸序列SISIAGSTYYNPSLKS(SEQ ID NO:26)所示的CDR H2、或所述CDR H2的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;和氨基酸序列DRGDQILDV(SEQ ID NO:27)所示的CDR H3、或所述CDR H3的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;所述轻链可变区包含根据Kabat编号的氨基酸序列RASQSISRYLN(SEQ ID NO:28)所示的CDR L1、或所述CDR L1的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;氨基酸序列AASSLQS(SEQ ID NO:29)所示的CDR L2、或所述CDR L2的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;和氨基酸序列QQKYFDIT(SEQ ID NO:30)所示的CDR L3、或所述CDR L3的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;(b)所述重链可变区包含根据Kabat编号的氨基酸序列NDVIS(SEQ ID NO:31)所示的CDR H1、或所述CDR H1的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;氨基酸序列VIIPIFGIANYAQKFQG(SEQ ID NO:32)所示的CDR H2、或所述CDR H2的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;和氨基酸序列GRGYYSSWLHDI(SEQ ID NO:33)所示的CDR H3、或所述CDR H3的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;所述轻链可变区包含根据Kabat编号的氨基酸序列QASQDITNYLN(SEQ ID NO:34)所示的CDR L1、或所述CDR L1的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;氨基酸序列DASNLET(SEQ ID NO:35)所示的CDR L2、或所述CDR L2的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;和氨基酸序列QQAFDLIT(SEQ ID NO:36)所示的CDR L3、或所述CDR L3的不超过2个氨基酸变化或不超过1个氨基酸变化的变体;其中所述氨基酸变化是氨基酸的添加、缺失或取代;例如,其中:(a)所述重链可变区包含根据Kabat编号的氨基酸序列SSSYYWT(SEQ ID NO:25)所示的CDR H1;氨基酸序列SISIAGSTYYNPSLKS(SEQ ID NO:26)所示的CDR H2;和氨基酸序列DRGDQILDV(SEQ ID NO:27)所示的CDR H3;所述轻链可变区包含根据Kabat编号的氨基酸序列RASQSISRYLN(SEQ ID NO:28)所示的CDR L1;氨基酸序列AASSLQS(SEQ ID NO:29)所示的CDR L2;和氨基酸序列QQKYFDIT(SEQ ID NO:30)所示的CDR L3;(b)所述重链可变区包含根据Kabat编号的氨基酸序列NDVIS(SEQ ID NO:31)所示的CDR H1;氨基酸序列VIIPIFGIANYAQKFQG(SEQ ID NO:32)所示的CDR H2;和氨基酸序列GRGYYSSWLHDI(SEQ ID NO:33)所示的CDR H3;所述轻链可变区包含根据Kabat编号的氨基酸序列QASQDITNYLN(SEQ ID NO:34)所示的CDR L1;氨基酸序列DASNLET(SEQ ID NO:35)所示的CDR L2;和氨基酸序列QQAFDLIT(SEQ ID NO:36)所示的CDR L3;例如,其中:(a)重链可变区包含SEQ ID NO:2的序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列,且轻链可变区包含SEQ ID NO:3的序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列;(b)重链可变区包含SEQ ID NO:9的序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列,且轻链可变区包含SEQ ID NO:10的序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列;例如,其中:(a)重链可变区包含SEQ ID NO:2的序列,且轻链可变区包含SEQ ID NO:3的序列;(b)重链可变区包含SEQ ID NO:9的序列,且轻链可变区包含SEQ ID NO:10的序列;例如,所述抗体是IgG1、IgG2、IgG3或IgG4抗体;优选地,其是IgG1或IgG4抗体;更优选地,其是IgG1抗体;例如,所述抗原结合片段是Fab、Fab’、F(ab’) 2、Fv、单链Fv、单链Fab、双体抗体(diabody)。
- 根据权利要求1或2所述的药物组合,其中所述突变Fc结构域是IgG1、IgG2、IgG3或IgG4抗体的突变Fc结构域,优选地,所述突变Fc结构域是IgG1或IgG4抗体的突变Fc结构域;更优选地,所述突变Fc结构域是IgG1抗体的突变Fc结构域;例如,所述抗体或抗原结合片段包含SEQ ID NO:5所示的重链恒定区序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列且其中根据EU编号的P329位置处的氨基酸突变为G;例如,所述抗体或抗原结合片段包含SEQ ID NO:5所示的重链恒定区序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列且其中根据EU编号的P329位置处的氨基酸突变为G;和SEQ ID NO:6所示的轻链恒定区序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列;例如,所述抗体或抗原结合片段包含SEQ ID NO:5所示的重链恒定区序列和SEQ ID NO:6所示的轻链恒定区序列。
- 根据权利要求1-3中任一项所述的药物组合,其中所述分子开关调控型CAR多肽还包含位于N端的信号肽序列,例如,SEQ ID NO:11所示的信号肽序列,优选地,所述分子开关调控型CAR多肽具有SEQ ID NO:1所示的氨基酸序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的序列。
- 根据权利要求1-4中任一项所述的药物组合,其中编码CAR多肽的核酸分子是编码权利要求1-4中任一项所述药物组合中所述的CAR多肽的核酸分子,例如,是编码SEQ ID NO:1所示的氨基酸序列或与其具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列的核酸分子。
- 根据权利要求1-4中任一项所述的药物组合,其中所述载体是包含编码权利要求1-4中任一所述药物组合中所述的CAR多肽的核酸分子的载体,例如,所述载体选自DNA载体、 RNA载体、质粒、慢病毒载体、腺病毒载体或逆转录病毒载体。
- 根据权利要求1-4中任一项所述的药物组合,其中所述免疫效应细胞是自自体T细胞、NK细胞或同种异体T细胞、NK细胞制备的表达权利要求1-4中任一项所述的药物组合中的分子开关调控型CAR多肽的T细胞、NK细胞,例如,所述免疫效应细胞是自人PBMC分离的T细胞、NK细胞制备的表达权利要求1-4中任一项所述的药物组合中的分子开关调控型CAR多肽的T细胞、NK细胞。
- 根据权利要求1-7中任一项所述的药物组合,其中(i)将表达权利要求1-4中任一项所述药物组合中所述的CAR多肽的免疫效应细胞以1×10 6个细胞/kg体重-10×10 6个细胞/kg体重,例如1×10 6个细胞/kg体重、2×10 6个细胞/kg体重、3×10 6个细胞/kg体重、5×10 6个细胞/kg体重、7×10 6个细胞/kg体重、9×10 6个细胞/kg体重、10×10 6个细胞/kg体重的剂量以单次或多次静脉内施用至受试者;和(ii)将权利要求1-4中任一项所述药物组合中所述的P329G突变抗体以0.1-10mg/kg、优选地0.1mg/kg、0.3mg/kg、0.5mg/kg、1mg/kg、3mg/kg、5mg/kg、7mg/kg、9mg/kg、10mg/kg的剂量单元的形式,优选地胃肠外、更优选地静脉内施用至受试者。
- 根据权利要求8所述的药物组合,其中(i)和(ii)分开、同时或依次施用,例如,第一天施用(ii),同一天静脉内施用(i),然后按照一定频率多次施用(ii),同时通过监测(i)的体内PK浓度和期望的治疗功效终点,确定是否多次施用(i);或者第一天施用(i),第二天静脉内施用(ii),然后按照一定频率多次施用(ii),同时通过监测(i)的体内PK浓度和期望的治疗功效终点,确定是否多次施用(i);或者第一天施用(ii),第二天静脉内施用(i),然后按照一定频率多次施用(ii),同时通过监测(i)的体内PK浓度和期望的治疗功效终点,确定是否多次施用(i);例如,(i)和(ii)各施用一次,然后每3-4天一次、每周一次、每两周一次、每三周一次或每四周一次的给药频率多次施用(ii),同时通过监测(i)的体内PK浓度和期望的治疗功效终点,确定是否多次施用(i)。
- 根据权利要求1-9中任一项所述的药物组合的用途,用于在受试者中治疗与BCMA相关的疾病,包括向受试者施用治疗有效量的权利要求1-9中任一项所述的药物组合,优选地,所述与BCMA相关的疾病是例如表达或过表达BCMA的癌症,所述癌症例如是复发/难治性多发性骨髓瘤(relapsed/refractory multiple myeloma,RRMM)。
- 根据权利要求1-9中任一项所述的药物组合在制备用于治疗与BCMA相关的疾病的药物中的用途,所述与BCMA相关的疾病是例如表达或过表达BCMA的癌症,所述癌症例如是复发/难治性多发性骨髓瘤(relapsed/refractory multiple myeloma,RRMM)。
- 用于治疗与BCMA相关的疾病的方法,所述方法包括向受试者施用治疗有效量的权利要求1-9中任一项所述的药物组合,所述疾病是例如表达或过表达BCMA的癌症,所述癌症例如是复发/难治性多发性骨髓瘤(relapsed/refractory multiple myeloma,RRMM)。
- 成套药盒,其包含权利要求1-9中任一项所述的药物组合,优选地所述药盒为药物剂量单元形式。
- 一种药物复合物,其是一种由(i)表达权利要求1-4中任一项所述药物组合中所述的分子开关调控型CAR多肽的免疫效应细胞(例如,T细胞、NK细胞);和(ii)包含P329G突变的特异性结合BCMA分子的抗体或抗原结合片段(也称为P329G突变抗体),例如,所述P329G突变抗体包含突变Fc结构域,其中根据EU编号的P329位置处的氨基酸突变为甘氨酸(G),与未突变的亲本抗体Fc结构域的Fcγ受体结合相比,突变Fc结构域的Fcγ受体结合降低;结合而产生的复合物,其中,所述免疫效应细胞通过CAR多肽的胞外结构域中的人源化抗P329G突变scFv序列与所述P329G突变抗体的Fc结构域结合而产生所述复合物;例如,其中所述免疫效应细胞是自自体T细胞、NK细胞或同种异体T细胞、NK细胞制备的表达权利要求1-4中任一项所述药物组合中所述的分子开关调控型CAR多肽的T细胞、NK细胞,例如,所述免疫效应细胞是自人PBMC分离的T细胞、NK细胞制备的表达权利要求1-4中任一项所述药物组合中所述的分子开关调控型CAR多肽的T细胞、NK细胞;例如,其中P329G突变抗体是ADI-38497 PG Ab和/或ADI-38484 PG Ab。
- 根据权利要求14所述的药物复合物的用途,用于在受试者中治疗与BCMA相关的疾病,优选地,所述与BCMA相关的疾病是例如表达或过表达BCMA的癌症,所述癌症例如是复发/难治性多发性骨髓瘤(relapsed/refractory multiple myeloma,RRMM)。
Priority Applications (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202280081143.2A CN118317789A (zh) | 2021-12-07 | 2022-12-07 | 靶向bcma的p329g抗体及其与嵌合抗原受体细胞的组合和应用 |
EP22903520.9A EP4445912A1 (en) | 2021-12-07 | 2022-12-07 | P329g antibody targeting bcma, combination of same with chimeric antigen receptor cell, and use thereof |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202111487287.1 | 2021-12-07 | ||
CN202111487287.1A CN116284385A (zh) | 2021-12-07 | 2021-12-07 | 靶向bcma的p329g抗体及其与嵌合抗原受体细胞的组合和应用 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2023104099A1 true WO2023104099A1 (zh) | 2023-06-15 |
Family
ID=86729651
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2022/137265 WO2023104099A1 (zh) | 2021-12-07 | 2022-12-07 | 靶向bcma的p329g抗体及其与嵌合抗原受体细胞的组合和应用 |
PCT/CN2022/137266 WO2023104100A1 (zh) | 2021-12-07 | 2022-12-07 | 结合bcma的抗体及其用途 |
Family Applications After (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2022/137266 WO2023104100A1 (zh) | 2021-12-07 | 2022-12-07 | 结合bcma的抗体及其用途 |
Country Status (6)
Country | Link |
---|---|
EP (2) | EP4445912A1 (zh) |
KR (1) | KR20240136952A (zh) |
CN (3) | CN116284385A (zh) |
AU (1) | AU2022405415A1 (zh) |
CA (1) | CA3241912A1 (zh) |
WO (2) | WO2023104099A1 (zh) |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN118477173A (zh) * | 2023-02-13 | 2024-08-13 | 信达细胞制药(苏州)有限公司 | 靶向HER2/p95HER2的P329G抗体及其与嵌合抗原受体细胞的组合和应用 |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9273141B2 (en) | 2011-05-27 | 2016-03-01 | Glaxo Group Limited | B cell maturation antigen (BCMA) binding proteins |
WO2019149269A1 (zh) * | 2018-02-01 | 2019-08-08 | 信达生物制药(苏州)有限公司 | 全人源的抗b细胞成熟抗原(bcma)单链抗体及其应用 |
CN110582298A (zh) * | 2017-02-17 | 2019-12-17 | 优努姆治疗公司 | 抗bcma抗体和抗体偶联t细胞受体(actr)在癌症治疗和b细胞疾病中的联合使用 |
CN110662560A (zh) * | 2017-03-27 | 2020-01-07 | 豪夫迈·罗氏有限公司 | 改进的抗原结合受体 |
US20200109202A1 (en) * | 2018-04-13 | 2020-04-09 | Affimed Gmbh | Nk cell engaging antibody fusion constructs |
WO2021121228A1 (en) * | 2019-12-16 | 2021-06-24 | Nanjing Legend Biotech Co., Ltd. | Single domain antibodies and chimeric antigen receptors targeting bcma and methods of use thereof |
US11066475B2 (en) * | 2017-11-01 | 2021-07-20 | Juno Therapeutics, Inc. | Chimeric antigen receptors specific for B-cell maturation antigen and encoding polynucleotides |
Family Cites Families (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5336603A (en) | 1987-10-02 | 1994-08-09 | Genentech, Inc. | CD4 adheson variants |
KR900005995A (ko) | 1988-10-31 | 1990-05-07 | 우메모또 요시마사 | 변형 인터류킨-2 및 그의 제조방법 |
US5622929A (en) | 1992-01-23 | 1997-04-22 | Bristol-Myers Squibb Company | Thioether conjugates |
CN105384825B (zh) * | 2015-08-11 | 2018-06-01 | 南京传奇生物科技有限公司 | 一种基于单域抗体的双特异性嵌合抗原受体及其应用 |
BR112019014986A8 (pt) * | 2017-01-23 | 2022-10-18 | Carsgen Therapeutics Co Ltd | Anticorpo que alveja bcma e uso do mesmo |
EP3777895A4 (en) * | 2018-06-26 | 2021-12-22 | ABL Bio Inc. | ANTI-BCMA ANTIBODIES AND USES THEREOF |
CN109265550B (zh) * | 2018-09-25 | 2020-09-15 | 华东师范大学 | Bcma抗体、嵌合抗原受体和药物 |
JP2023501871A (ja) * | 2019-09-20 | 2023-01-20 | 上海吉倍生物技術有限公司 | Bcma標的化抗体及びキメラ抗原受容体 |
WO2021146147A1 (en) * | 2020-01-13 | 2021-07-22 | Nkarta, Inc. | Bcma-directed cellular immunotherapy compositions and methods |
-
2021
- 2021-12-07 CN CN202111487287.1A patent/CN116284385A/zh active Pending
-
2022
- 2022-12-07 EP EP22903520.9A patent/EP4445912A1/en active Pending
- 2022-12-07 CA CA3241912A patent/CA3241912A1/en active Pending
- 2022-12-07 WO PCT/CN2022/137265 patent/WO2023104099A1/zh active Application Filing
- 2022-12-07 KR KR1020247022135A patent/KR20240136952A/ko unknown
- 2022-12-07 WO PCT/CN2022/137266 patent/WO2023104100A1/zh active Application Filing
- 2022-12-07 AU AU2022405415A patent/AU2022405415A1/en active Pending
- 2022-12-07 CN CN202280081161.0A patent/CN118339185A/zh active Pending
- 2022-12-07 EP EP22903521.7A patent/EP4446341A1/en active Pending
- 2022-12-07 CN CN202280081143.2A patent/CN118317789A/zh active Pending
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9273141B2 (en) | 2011-05-27 | 2016-03-01 | Glaxo Group Limited | B cell maturation antigen (BCMA) binding proteins |
CN110582298A (zh) * | 2017-02-17 | 2019-12-17 | 优努姆治疗公司 | 抗bcma抗体和抗体偶联t细胞受体(actr)在癌症治疗和b细胞疾病中的联合使用 |
CN110662560A (zh) * | 2017-03-27 | 2020-01-07 | 豪夫迈·罗氏有限公司 | 改进的抗原结合受体 |
US11066475B2 (en) * | 2017-11-01 | 2021-07-20 | Juno Therapeutics, Inc. | Chimeric antigen receptors specific for B-cell maturation antigen and encoding polynucleotides |
WO2019149269A1 (zh) * | 2018-02-01 | 2019-08-08 | 信达生物制药(苏州)有限公司 | 全人源的抗b细胞成熟抗原(bcma)单链抗体及其应用 |
US20200109202A1 (en) * | 2018-04-13 | 2020-04-09 | Affimed Gmbh | Nk cell engaging antibody fusion constructs |
WO2021121228A1 (en) * | 2019-12-16 | 2021-06-24 | Nanjing Legend Biotech Co., Ltd. | Single domain antibodies and chimeric antigen receptors targeting bcma and methods of use thereof |
Non-Patent Citations (17)
Title |
---|
"GenBank", Database accession no. AAA62478.2 |
"Remington's Pharmaceutical Science", MARK PUBLISHING COMPANY |
"Sorting Out Fluorescence Activated Cell Sorting", FLOWMETRIC, 9 November 2017 (2017-11-09) |
AL-LAZIKANI ET AL.: "Standard conformations for the canonical structures of immunoglobulins", JOURNAL OF MOLECULAR BIOLOGY, vol. 273, 1997, pages 927 - 948, XP004461383, DOI: 10.1006/jmbi.1997.1354 |
ARMOUR ET AL., J. IMMUNOL., vol. 29, no. 8, 1999, pages 2613 - 2624 |
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 877 - 883 |
E. MEYERSW. MILLER, CABIOS, vol. 4, 1989, pages 11 - 17 |
FENG, DEMING ET AL.: "Overview of Anti-BCMA CAR-T Immunotherapy for Multiple Myeloma and Relapsed/Refractory Multiple Myeloma", SCANDINAVIAN JOURNAL OF IMMUNOLOGY, 31 December 2020 (2020-12-31), XP055881933 * |
FLATMAN ET AL., J. CHROMATOGR., vol. B848, 2007, pages 79 - 87 |
GAZZANO-SANTORO ET AL., J. IMMUNOL. METHODS, vol. 202, 1996, pages 163 |
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1987, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES |
KENDERIAN SS ET AL.: "CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia[J", LEUKEMIA, vol. 29, no. 8, 2015, pages 1637 - 1647, XP055227771, DOI: 10.1038/leu.2015.52 |
MILONE ET AL., MOL. THER., vol. 17, no. 8, 2009, pages 1453 - 1464 |
NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 444 - 453 |
SHIELDS ET AL., J. BIOL. CHEM, vol. 276, no. 9, 2001, pages 6591 - 6604 |
YAZAKIWU: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, pages: 255 - 268 |
ZHANG HUIHUI ET AL.: "Preclinical research on suicide gene as ''safety switch'' to control CAR-T cell toxicity", CHINESE JOURNAL OF CANCER BIOTHERAPY, vol. 28, no. 3, 2021, pages 225 - 231 |
Also Published As
Publication number | Publication date |
---|---|
KR20240136952A (ko) | 2024-09-19 |
CN118317789A (zh) | 2024-07-09 |
WO2023104100A1 (zh) | 2023-06-15 |
CN116284385A (zh) | 2023-06-23 |
AU2022405415A1 (en) | 2024-07-18 |
CN118339185A (zh) | 2024-07-12 |
CA3241912A1 (en) | 2023-06-15 |
EP4445912A1 (en) | 2024-10-16 |
EP4446341A1 (en) | 2024-10-16 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7386382B2 (ja) | キメラ抗原受容体及びt細胞受容体並びに使用方法 | |
US20240092855A1 (en) | Engineered proteins to enhance sensitivity of a cell to il-2 | |
EP3875484A1 (en) | Cll1-targeting antibody and application thereof | |
AU2020207962A1 (en) | Methods and compositions to improve the safety and efficacy of cellular therapies | |
CN110950953A (zh) | 抗b7-h3的单克隆抗体及其在细胞治疗中的应用 | |
US20230203178A1 (en) | Chimeric antigen receptor car or car construct targeting bcma and cd19 and application thereof | |
WO2021043169A1 (zh) | 特异性结合b细胞成熟抗原的抗体及其用途 | |
WO2023020459A1 (zh) | 靶向SIRPα的单克隆抗体及其用途 | |
WO2023093811A1 (zh) | 分子开关调控型嵌合抗原受体细胞和抗体的组合及其应用 | |
TW202116812A (zh) | 抗dll3嵌合抗原受體及其用途 | |
CA2968987C (en) | Soluble universal adcc-enhancing synthetic fusion gene and peptide technology and its use thereof | |
CN114302733A (zh) | 抗纽约食管鳞状细胞癌1(ny-eso-1)抗原结合蛋白及其使用方法 | |
WO2023104099A1 (zh) | 靶向bcma的p329g抗体及其与嵌合抗原受体细胞的组合和应用 | |
WO2023160260A1 (zh) | Cd7-car-t细胞及其制备方法和应用 | |
WO2023030539A1 (en) | Anti-gpc3 chimeric antigen receptor and methods of use thereof | |
WO2023173272A1 (zh) | 靶向gprc5d的全人源嵌合抗原受体(car)及其应用 | |
WO2024230776A1 (zh) | Bcmap329g抗体及car-t细胞在治疗多发性骨髓瘤中的应用 | |
WO2024140836A1 (zh) | 靶向nkg2d配体的融合蛋白与嵌合抗原受体细胞的组合及其用途 | |
WO2024169985A1 (zh) | 靶向HER2/p95HER2的P329G抗体及其与嵌合抗原受体细胞的组合和应用 | |
CN118924893A (zh) | Bcmap329g抗体及car-t细胞在治疗多发性骨髓瘤中的应用 | |
WO2024088371A1 (zh) | 靶向msln的抗原结合蛋白 | |
CN114933654B (zh) | 靶向cd123的抗体、嵌合抗原受体及其用途 | |
US20240343805A1 (en) | USE OF AN ANTI-TGFßR2/PD-1 BISPECIFIC ANTIBODY | |
WO2024103251A1 (zh) | 抗afp/hla02 tcr样抗体及其用途 | |
WO2024082178A1 (zh) | 靶向cd19和cd22的双特异性嵌合抗原受体 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 22903520 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 202280081143.2 Country of ref document: CN |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2022903520 Country of ref document: EP |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2022903520 Country of ref document: EP Effective date: 20240708 |