CN118139897A - Synthetic T cell receptor antigen receptor specifically combined with mesothelin and application thereof - Google Patents
Synthetic T cell receptor antigen receptor specifically combined with mesothelin and application thereof Download PDFInfo
- Publication number
- CN118139897A CN118139897A CN202280071907.XA CN202280071907A CN118139897A CN 118139897 A CN118139897 A CN 118139897A CN 202280071907 A CN202280071907 A CN 202280071907A CN 118139897 A CN118139897 A CN 118139897A
- Authority
- CN
- China
- Prior art keywords
- constant region
- tcr
- amino acid
- chain constant
- chain
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 title claims abstract description 323
- 108090000015 Mesothelin Proteins 0.000 title claims abstract description 75
- 102000003735 Mesothelin Human genes 0.000 title claims description 74
- 239000000427 antigen Substances 0.000 claims abstract description 147
- 108091007433 antigens Proteins 0.000 claims abstract description 147
- 102000036639 antigens Human genes 0.000 claims abstract description 147
- 230000027455 binding Effects 0.000 claims abstract description 137
- 239000012634 fragment Substances 0.000 claims abstract description 65
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 52
- 150000001413 amino acids Chemical group 0.000 claims description 186
- 235000001014 amino acid Nutrition 0.000 claims description 172
- 229940024606 amino acid Drugs 0.000 claims description 171
- 210000004027 cell Anatomy 0.000 claims description 167
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 145
- 241001529936 Murinae Species 0.000 claims description 127
- 230000003834 intracellular effect Effects 0.000 claims description 106
- 241000282414 Homo sapiens Species 0.000 claims description 66
- 235000018417 cysteine Nutrition 0.000 claims description 54
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 claims description 53
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Natural products NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 claims description 44
- 108010003723 Single-Domain Antibodies Proteins 0.000 claims description 43
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 43
- 229920001184 polypeptide Polymers 0.000 claims description 37
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 37
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 claims description 36
- 230000035772 mutation Effects 0.000 claims description 32
- 150000007523 nucleic acids Chemical class 0.000 claims description 31
- 230000004048 modification Effects 0.000 claims description 29
- 238000012986 modification Methods 0.000 claims description 29
- 102000005962 receptors Human genes 0.000 claims description 28
- 108020003175 receptors Proteins 0.000 claims description 28
- 210000002865 immune cell Anatomy 0.000 claims description 27
- 238000000034 method Methods 0.000 claims description 27
- 102000039446 nucleic acids Human genes 0.000 claims description 26
- 108020004707 nucleic acids Proteins 0.000 claims description 26
- 108090000623 proteins and genes Proteins 0.000 claims description 25
- 102000006306 Antigen Receptors Human genes 0.000 claims description 24
- 239000004471 Glycine Substances 0.000 claims description 24
- 108010083359 Antigen Receptors Proteins 0.000 claims description 22
- -1 ICOS Proteins 0.000 claims description 22
- 102000004169 proteins and genes Human genes 0.000 claims description 22
- 239000013598 vector Substances 0.000 claims description 22
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 claims description 21
- 239000004473 Threonine Substances 0.000 claims description 21
- 230000037430 deletion Effects 0.000 claims description 21
- 238000012217 deletion Methods 0.000 claims description 21
- 230000014509 gene expression Effects 0.000 claims description 21
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 claims description 20
- 229930182817 methionine Natural products 0.000 claims description 20
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 claims description 19
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 claims description 19
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 claims description 19
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 claims description 19
- 210000004899 c-terminal region Anatomy 0.000 claims description 19
- 102000003675 cytokine receptors Human genes 0.000 claims description 19
- 108010057085 cytokine receptors Proteins 0.000 claims description 19
- 235000018102 proteins Nutrition 0.000 claims description 19
- 239000004474 valine Substances 0.000 claims description 19
- 239000002773 nucleotide Substances 0.000 claims description 18
- 125000003729 nucleotide group Chemical group 0.000 claims description 18
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 claims description 17
- 229960000310 isoleucine Drugs 0.000 claims description 17
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 claims description 17
- 239000004475 Arginine Substances 0.000 claims description 15
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 claims description 15
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 claims description 15
- 239000004472 Lysine Substances 0.000 claims description 15
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 claims description 15
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 claims description 15
- 239000013604 expression vector Substances 0.000 claims description 13
- 210000004986 primary T-cell Anatomy 0.000 claims description 11
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 claims description 10
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 claims description 10
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 9
- 125000000637 arginyl group Chemical group N[C@@H](CCCNC(N)=N)C(=O)* 0.000 claims description 9
- 101150013553 CD40 gene Proteins 0.000 claims description 8
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 claims description 8
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 claims description 8
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 claims description 8
- 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 claims description 8
- 229910052700 potassium Inorganic materials 0.000 claims description 8
- 201000011510 cancer Diseases 0.000 claims description 7
- 238000002360 preparation method Methods 0.000 claims description 7
- 102100027207 CD27 antigen Human genes 0.000 claims description 6
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 claims description 6
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 6
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims description 6
- 206010039491 Sarcoma Diseases 0.000 claims description 6
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 claims description 6
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 claims description 6
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 claims description 6
- 208000032839 leukemia Diseases 0.000 claims description 6
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims description 6
- 201000002528 pancreatic cancer Diseases 0.000 claims description 6
- 208000008443 pancreatic carcinoma Diseases 0.000 claims description 6
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 claims description 6
- 239000008194 pharmaceutical composition Substances 0.000 claims description 6
- 108010052285 Membrane Proteins Proteins 0.000 claims description 5
- 238000012258 culturing Methods 0.000 claims description 5
- 210000004443 dendritic cell Anatomy 0.000 claims description 5
- 206010006187 Breast cancer Diseases 0.000 claims description 4
- 208000026310 Breast neoplasm Diseases 0.000 claims description 4
- 206010025323 Lymphomas Diseases 0.000 claims description 4
- 108010033276 Peptide Fragments Proteins 0.000 claims description 4
- 102000007079 Peptide Fragments Human genes 0.000 claims description 4
- 206010060862 Prostate cancer Diseases 0.000 claims description 4
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 4
- 208000005718 Stomach Neoplasms Diseases 0.000 claims description 4
- 238000003745 diagnosis Methods 0.000 claims description 4
- 206010017758 gastric cancer Diseases 0.000 claims description 4
- 125000003630 glycyl group Chemical group [H]N([H])C([H])([H])C(*)=O 0.000 claims description 4
- 210000000822 natural killer cell Anatomy 0.000 claims description 4
- 201000011549 stomach cancer Diseases 0.000 claims description 4
- 102100026882 Alpha-synuclein Human genes 0.000 claims description 3
- 206010009944 Colon cancer Diseases 0.000 claims description 3
- 108010003384 Colony-Stimulating Factor Receptors Proteins 0.000 claims description 3
- 102000004626 Colony-Stimulating Factor Receptors Human genes 0.000 claims description 3
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 claims description 3
- 206010014733 Endometrial cancer Diseases 0.000 claims description 3
- 206010014759 Endometrial neoplasm Diseases 0.000 claims description 3
- 208000000461 Esophageal Neoplasms Diseases 0.000 claims description 3
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 claims description 3
- 101000834898 Homo sapiens Alpha-synuclein Proteins 0.000 claims description 3
- 101000889276 Homo sapiens Cytotoxic T-lymphocyte protein 4 Proteins 0.000 claims description 3
- 101001068133 Homo sapiens Hepatitis A virus cellular receptor 2 Proteins 0.000 claims description 3
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 claims description 3
- 101000611936 Homo sapiens Programmed cell death protein 1 Proteins 0.000 claims description 3
- 101000652359 Homo sapiens Spermatogenesis-associated protein 2 Proteins 0.000 claims description 3
- 102000001617 Interferon Receptors Human genes 0.000 claims description 3
- 108010054267 Interferon Receptors Proteins 0.000 claims description 3
- 102000017578 LAG3 Human genes 0.000 claims description 3
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 3
- 206010030155 Oesophageal carcinoma Diseases 0.000 claims description 3
- 206010033128 Ovarian cancer Diseases 0.000 claims description 3
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 3
- 208000006990 cholangiocarcinoma Diseases 0.000 claims description 3
- 208000029742 colonic neoplasm Diseases 0.000 claims description 3
- 201000004101 esophageal cancer Diseases 0.000 claims description 3
- 230000001939 inductive effect Effects 0.000 claims description 3
- 108010093036 interleukin receptors Proteins 0.000 claims description 3
- 102000002467 interleukin receptors Human genes 0.000 claims description 3
- 210000004185 liver Anatomy 0.000 claims description 3
- 201000005202 lung cancer Diseases 0.000 claims description 3
- 208000020816 lung neoplasm Diseases 0.000 claims description 3
- 210000002540 macrophage Anatomy 0.000 claims description 3
- 206010042863 synovial sarcoma Diseases 0.000 claims description 3
- 230000001131 transforming effect Effects 0.000 claims description 3
- 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 2
- 208000031261 Acute myeloid leukaemia Diseases 0.000 claims description 2
- 201000003076 Angiosarcoma Diseases 0.000 claims description 2
- 206010003571 Astrocytoma Diseases 0.000 claims description 2
- 208000036170 B-Cell Marginal Zone Lymphoma Diseases 0.000 claims description 2
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 claims description 2
- 208000003950 B-cell lymphoma Diseases 0.000 claims description 2
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 claims description 2
- 206010004593 Bile duct cancer Diseases 0.000 claims description 2
- 206010005003 Bladder cancer Diseases 0.000 claims description 2
- 206010005949 Bone cancer Diseases 0.000 claims description 2
- 208000018084 Bone neoplasm Diseases 0.000 claims description 2
- 208000005243 Chondrosarcoma Diseases 0.000 claims description 2
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 claims description 2
- 208000006168 Ewing Sarcoma Diseases 0.000 claims description 2
- 201000008808 Fibrosarcoma Diseases 0.000 claims description 2
- 208000001258 Hemangiosarcoma Diseases 0.000 claims description 2
- 208000017604 Hodgkin disease Diseases 0.000 claims description 2
- 208000021519 Hodgkin lymphoma Diseases 0.000 claims description 2
- 208000010747 Hodgkins lymphoma Diseases 0.000 claims description 2
- 208000008839 Kidney Neoplasms Diseases 0.000 claims description 2
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 claims description 2
- 208000018142 Leiomyosarcoma Diseases 0.000 claims description 2
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 claims description 2
- 208000025205 Mantle-Cell Lymphoma Diseases 0.000 claims description 2
- 208000034578 Multiple myelomas Diseases 0.000 claims description 2
- 201000003793 Myelodysplastic syndrome Diseases 0.000 claims description 2
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 claims description 2
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 claims description 2
- 208000001894 Nasopharyngeal Neoplasms Diseases 0.000 claims description 2
- 206010061306 Nasopharyngeal cancer Diseases 0.000 claims description 2
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims description 2
- 206010035226 Plasma cell myeloma Diseases 0.000 claims description 2
- 208000015634 Rectal Neoplasms Diseases 0.000 claims description 2
- 206010038389 Renal cancer Diseases 0.000 claims description 2
- 208000021712 Soft tissue sarcoma Diseases 0.000 claims description 2
- 206010042971 T-cell lymphoma Diseases 0.000 claims description 2
- 208000027585 T-cell non-Hodgkin lymphoma Diseases 0.000 claims description 2
- 208000024313 Testicular Neoplasms Diseases 0.000 claims description 2
- 206010057644 Testis cancer Diseases 0.000 claims description 2
- 208000024770 Thyroid neoplasm Diseases 0.000 claims description 2
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 claims description 2
- 208000033559 Waldenström macroglobulinemia Diseases 0.000 claims description 2
- 230000001154 acute effect Effects 0.000 claims description 2
- 208000026900 bile duct neoplasm Diseases 0.000 claims description 2
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 claims description 2
- 206010012818 diffuse large B-cell lymphoma Diseases 0.000 claims description 2
- 201000003444 follicular lymphoma Diseases 0.000 claims description 2
- 208000005017 glioblastoma Diseases 0.000 claims description 2
- 201000010536 head and neck cancer Diseases 0.000 claims description 2
- 208000014829 head and neck neoplasm Diseases 0.000 claims description 2
- 201000010982 kidney cancer Diseases 0.000 claims description 2
- 206010024627 liposarcoma Diseases 0.000 claims description 2
- 201000007270 liver cancer Diseases 0.000 claims description 2
- 208000014018 liver neoplasm Diseases 0.000 claims description 2
- 201000007924 marginal zone B-cell lymphoma Diseases 0.000 claims description 2
- 208000021937 marginal zone lymphoma Diseases 0.000 claims description 2
- 201000001441 melanoma Diseases 0.000 claims description 2
- 210000000581 natural killer T-cell Anatomy 0.000 claims description 2
- 208000002154 non-small cell lung carcinoma Diseases 0.000 claims description 2
- 201000008968 osteosarcoma Diseases 0.000 claims description 2
- 208000031223 plasma cell leukemia Diseases 0.000 claims description 2
- 206010038038 rectal cancer Diseases 0.000 claims description 2
- 201000001275 rectum cancer Diseases 0.000 claims description 2
- 210000003289 regulatory T cell Anatomy 0.000 claims description 2
- 201000009410 rhabdomyosarcoma Diseases 0.000 claims description 2
- 238000006467 substitution reaction Methods 0.000 claims description 2
- 201000003120 testicular cancer Diseases 0.000 claims description 2
- 201000002510 thyroid cancer Diseases 0.000 claims description 2
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 claims description 2
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 claims description 2
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 2
- 230000000139 costimulatory effect Effects 0.000 claims 2
- 102000003390 tumor necrosis factor Human genes 0.000 claims 1
- 230000000694 effects Effects 0.000 abstract description 15
- 230000008685 targeting Effects 0.000 abstract description 7
- 102100025096 Mesothelin Human genes 0.000 abstract description 3
- 101000576802 Homo sapiens Mesothelin Proteins 0.000 abstract description 2
- 108091008874 T cell receptors Proteins 0.000 description 252
- 241000699666 Mus <mouse, genus> Species 0.000 description 150
- 125000003275 alpha amino acid group Chemical group 0.000 description 96
- 241000283984 Rodentia Species 0.000 description 95
- 230000002147 killing effect Effects 0.000 description 31
- 241000699670 Mus sp. Species 0.000 description 28
- 239000006228 supernatant Substances 0.000 description 26
- 239000000047 product Substances 0.000 description 24
- 239000000203 mixture Substances 0.000 description 22
- 230000006870 function Effects 0.000 description 21
- 239000013612 plasmid Substances 0.000 description 21
- 239000000243 solution Substances 0.000 description 21
- 238000001514 detection method Methods 0.000 description 20
- 241000700605 Viruses Species 0.000 description 19
- 238000002965 ELISA Methods 0.000 description 15
- 108010002350 Interleukin-2 Proteins 0.000 description 15
- 102000000588 Interleukin-2 Human genes 0.000 description 15
- 241001416177 Vicugna pacos Species 0.000 description 15
- 238000001727 in vivo Methods 0.000 description 15
- 239000002609 medium Substances 0.000 description 15
- 208000015181 infectious disease Diseases 0.000 description 14
- 239000007788 liquid Substances 0.000 description 14
- 238000005406 washing Methods 0.000 description 14
- 102000004127 Cytokines Human genes 0.000 description 13
- 108090000695 Cytokines Proteins 0.000 description 13
- 230000002209 hydrophobic effect Effects 0.000 description 13
- 238000000338 in vitro Methods 0.000 description 13
- 230000035755 proliferation Effects 0.000 description 13
- 230000004913 activation Effects 0.000 description 12
- 238000013461 design Methods 0.000 description 12
- 102100037850 Interferon gamma Human genes 0.000 description 11
- 108010074328 Interferon-gamma Proteins 0.000 description 11
- 238000003501 co-culture Methods 0.000 description 11
- 238000012216 screening Methods 0.000 description 11
- 238000011191 terminal modification Methods 0.000 description 11
- 230000022534 cell killing Effects 0.000 description 10
- 239000001963 growth medium Substances 0.000 description 10
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 9
- 239000002671 adjuvant Substances 0.000 description 9
- 230000001580 bacterial effect Effects 0.000 description 9
- 238000011161 development Methods 0.000 description 9
- 230000018109 developmental process Effects 0.000 description 9
- UQLDLKMNUJERMK-UHFFFAOYSA-L di(octadecanoyloxy)lead Chemical compound [Pb+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O UQLDLKMNUJERMK-UHFFFAOYSA-L 0.000 description 9
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 9
- 210000003071 memory t lymphocyte Anatomy 0.000 description 9
- 230000028327 secretion Effects 0.000 description 9
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 9
- 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 description 8
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 8
- 102100033726 Tumor necrosis factor receptor superfamily member 17 Human genes 0.000 description 8
- 230000008859 change Effects 0.000 description 8
- 238000006243 chemical reaction Methods 0.000 description 8
- 238000002474 experimental method Methods 0.000 description 8
- 230000003053 immunization Effects 0.000 description 8
- 238000002649 immunization Methods 0.000 description 8
- 210000001519 tissue Anatomy 0.000 description 8
- 108020004414 DNA Proteins 0.000 description 7
- 210000004369 blood Anatomy 0.000 description 7
- 239000008280 blood Substances 0.000 description 7
- 201000010099 disease Diseases 0.000 description 7
- 239000012636 effector Substances 0.000 description 7
- 239000011148 porous material Substances 0.000 description 7
- 239000003755 preservative agent Substances 0.000 description 7
- 230000002335 preservative effect Effects 0.000 description 7
- 230000005909 tumor killing Effects 0.000 description 7
- 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 6
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 6
- 102000001301 EGF receptor Human genes 0.000 description 6
- 108060006698 EGF receptor Proteins 0.000 description 6
- 101000801255 Homo sapiens Tumor necrosis factor receptor superfamily member 17 Proteins 0.000 description 6
- 102100034872 Kallikrein-4 Human genes 0.000 description 6
- 108060001084 Luciferase Proteins 0.000 description 6
- 239000005089 Luciferase Substances 0.000 description 6
- 241001465754 Metazoa Species 0.000 description 6
- 230000006044 T cell activation Effects 0.000 description 6
- 102000013529 alpha-Fetoproteins Human genes 0.000 description 6
- 108010026331 alpha-Fetoproteins Proteins 0.000 description 6
- 230000024245 cell differentiation Effects 0.000 description 6
- 238000002955 isolation Methods 0.000 description 6
- 108010024383 kallikrein 4 Proteins 0.000 description 6
- 239000012528 membrane Substances 0.000 description 6
- 238000007789 sealing Methods 0.000 description 6
- 210000002966 serum Anatomy 0.000 description 6
- 230000000638 stimulation Effects 0.000 description 6
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 5
- 230000003213 activating effect Effects 0.000 description 5
- 238000004364 calculation method Methods 0.000 description 5
- 238000010276 construction Methods 0.000 description 5
- 238000011534 incubation Methods 0.000 description 5
- 239000006166 lysate Substances 0.000 description 5
- 238000002156 mixing Methods 0.000 description 5
- 238000005457 optimization Methods 0.000 description 5
- 239000008188 pellet Substances 0.000 description 5
- 210000004881 tumor cell Anatomy 0.000 description 5
- 108010022366 Carcinoembryonic Antigen Proteins 0.000 description 4
- 102100025475 Carcinoembryonic antigen-related cell adhesion molecule 5 Human genes 0.000 description 4
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 4
- 102000018651 Epithelial Cell Adhesion Molecule Human genes 0.000 description 4
- 108010066687 Epithelial Cell Adhesion Molecule Proteins 0.000 description 4
- 102000016359 Fibronectins Human genes 0.000 description 4
- 108010067306 Fibronectins Proteins 0.000 description 4
- 101000874179 Homo sapiens Syndecan-1 Proteins 0.000 description 4
- 101000835093 Homo sapiens Transferrin receptor protein 1 Proteins 0.000 description 4
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 4
- 241000283973 Oryctolagus cuniculus Species 0.000 description 4
- 229920002873 Polyethylenimine Polymers 0.000 description 4
- 229920001213 Polysorbate 20 Polymers 0.000 description 4
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 4
- 102100035721 Syndecan-1 Human genes 0.000 description 4
- 230000006052 T cell proliferation Effects 0.000 description 4
- 102100026144 Transferrin receptor protein 1 Human genes 0.000 description 4
- 230000037396 body weight Effects 0.000 description 4
- 230000004069 differentiation Effects 0.000 description 4
- 239000003814 drug Substances 0.000 description 4
- 238000000605 extraction Methods 0.000 description 4
- 238000000684 flow cytometry Methods 0.000 description 4
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 4
- 230000031700 light absorption Effects 0.000 description 4
- 239000013642 negative control Substances 0.000 description 4
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 4
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 238000012163 sequencing technique Methods 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- 230000004083 survival effect Effects 0.000 description 4
- HBZBAMXERPYTFS-SECBINFHSA-N (4S)-2-(6,7-dihydro-5H-pyrrolo[3,2-f][1,3]benzothiazol-2-yl)-4,5-dihydro-1,3-thiazole-4-carboxylic acid Chemical compound OC(=O)[C@H]1CSC(=N1)c1nc2cc3CCNc3cc2s1 HBZBAMXERPYTFS-SECBINFHSA-N 0.000 description 3
- 241000282693 Cercopithecidae Species 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 229920001917 Ficoll Polymers 0.000 description 3
- 101000599940 Homo sapiens Interferon gamma Proteins 0.000 description 3
- 101100534458 Homo sapiens STAT5A gene Proteins 0.000 description 3
- 101000611183 Homo sapiens Tumor necrosis factor Proteins 0.000 description 3
- 108010038453 Interleukin-2 Receptors Proteins 0.000 description 3
- 102000010789 Interleukin-2 Receptors Human genes 0.000 description 3
- 101100096028 Mus musculus Smok1 gene Proteins 0.000 description 3
- 108091081024 Start codon Proteins 0.000 description 3
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 230000003915 cell function Effects 0.000 description 3
- 210000000170 cell membrane Anatomy 0.000 description 3
- 239000011248 coating agent Substances 0.000 description 3
- 238000000576 coating method Methods 0.000 description 3
- 230000000052 comparative effect Effects 0.000 description 3
- 239000012228 culture supernatant Substances 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 239000012895 dilution Substances 0.000 description 3
- 239000003480 eluent Substances 0.000 description 3
- 230000004927 fusion Effects 0.000 description 3
- 238000001502 gel electrophoresis Methods 0.000 description 3
- 102000057041 human TNF Human genes 0.000 description 3
- 230000005917 in vivo anti-tumor Effects 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 238000011081 inoculation Methods 0.000 description 3
- 230000004068 intracellular signaling Effects 0.000 description 3
- 239000010410 layer Substances 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 239000011259 mixed solution Substances 0.000 description 3
- 238000004806 packaging method and process Methods 0.000 description 3
- 244000052769 pathogen Species 0.000 description 3
- 230000002688 persistence Effects 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 238000010839 reverse transcription Methods 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 238000012546 transfer Methods 0.000 description 3
- 210000003462 vein Anatomy 0.000 description 3
- 230000003612 virological effect Effects 0.000 description 3
- 239000011534 wash buffer Substances 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- 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 description 2
- 241000714175 Abelson murine leukemia virus Species 0.000 description 2
- 101710145634 Antigen 1 Proteins 0.000 description 2
- 101100279855 Arabidopsis thaliana EPFL5 gene Proteins 0.000 description 2
- 101100520452 Arabidopsis thaliana PMD2 gene Proteins 0.000 description 2
- 241000713838 Avian myeloblastosis virus Species 0.000 description 2
- 108091008875 B cell receptors Proteins 0.000 description 2
- 108010008014 B-Cell Maturation Antigen Proteins 0.000 description 2
- 102100038080 B-cell receptor CD22 Human genes 0.000 description 2
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 2
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 102100026094 C-type lectin domain family 12 member A Human genes 0.000 description 2
- 101150031358 COLEC10 gene Proteins 0.000 description 2
- 101100314454 Caenorhabditis elegans tra-1 gene Proteins 0.000 description 2
- 102100025570 Cancer/testis antigen 1 Human genes 0.000 description 2
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 2
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 102100023721 Ephrin-B2 Human genes 0.000 description 2
- 108010044090 Ephrin-B2 Proteins 0.000 description 2
- 241000713730 Equine infectious anemia virus Species 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- 102100031507 Fc receptor-like protein 5 Human genes 0.000 description 2
- 101150032879 Fcrl5 gene Proteins 0.000 description 2
- 241000714475 Fujinami sarcoma virus Species 0.000 description 2
- 102100021197 G-protein coupled receptor family C group 5 member D Human genes 0.000 description 2
- 208000032612 Glial tumor Diseases 0.000 description 2
- 206010018338 Glioma Diseases 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 102100041003 Glutamate carboxypeptidase 2 Human genes 0.000 description 2
- 102100032530 Glypican-3 Human genes 0.000 description 2
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 2
- 102100028113 Granulocyte-macrophage colony-stimulating factor receptor subunit alpha Human genes 0.000 description 2
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 2
- 102100026122 High affinity immunoglobulin gamma Fc receptor I Human genes 0.000 description 2
- 101000884305 Homo sapiens B-cell receptor CD22 Proteins 0.000 description 2
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 2
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 2
- 101100496086 Homo sapiens CLEC12A gene Proteins 0.000 description 2
- 101000856237 Homo sapiens Cancer/testis antigen 1 Proteins 0.000 description 2
- 101000721661 Homo sapiens Cellular tumor antigen p53 Proteins 0.000 description 2
- 101001040713 Homo sapiens G-protein coupled receptor family C group 5 member D Proteins 0.000 description 2
- 101000892862 Homo sapiens Glutamate carboxypeptidase 2 Proteins 0.000 description 2
- 101001014668 Homo sapiens Glypican-3 Proteins 0.000 description 2
- 101000916625 Homo sapiens Granulocyte-macrophage colony-stimulating factor receptor subunit alpha Proteins 0.000 description 2
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 2
- 101000913074 Homo sapiens High affinity immunoglobulin gamma Fc receptor I Proteins 0.000 description 2
- 101001103039 Homo sapiens Inactive tyrosine-protein kinase transmembrane receptor ROR1 Proteins 0.000 description 2
- 101000599951 Homo sapiens Insulin-like growth factor I Proteins 0.000 description 2
- 101000994322 Homo sapiens Integrin alpha-8 Proteins 0.000 description 2
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 2
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 2
- 101000998120 Homo sapiens Interleukin-3 receptor subunit alpha Proteins 0.000 description 2
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 2
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 2
- 101001014223 Homo sapiens MAPK/MAK/MRK overlapping kinase Proteins 0.000 description 2
- 101001008874 Homo sapiens Mast/stem cell growth factor receptor Kit Proteins 0.000 description 2
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 2
- 101001109501 Homo sapiens NKG2-D type II integral membrane protein Proteins 0.000 description 2
- 101001103036 Homo sapiens Nuclear receptor ROR-alpha Proteins 0.000 description 2
- 101000891028 Homo sapiens Peptidyl-prolyl cis-trans isomerase FKBP11 Proteins 0.000 description 2
- 101000610551 Homo sapiens Prominin-1 Proteins 0.000 description 2
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 2
- 101000633784 Homo sapiens SLAM family member 7 Proteins 0.000 description 2
- 101000884271 Homo sapiens Signal transducer CD24 Proteins 0.000 description 2
- 101000716102 Homo sapiens T-cell surface glycoprotein CD4 Proteins 0.000 description 2
- 101000946843 Homo sapiens T-cell surface glycoprotein CD8 alpha chain Proteins 0.000 description 2
- 101000596234 Homo sapiens T-cell surface protein tactile Proteins 0.000 description 2
- 101000655352 Homo sapiens Telomerase reverse transcriptase Proteins 0.000 description 2
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 101150088952 IGF1 gene Proteins 0.000 description 2
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 2
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 2
- 102100039615 Inactive tyrosine-protein kinase transmembrane receptor ROR1 Human genes 0.000 description 2
- 102000004877 Insulin Human genes 0.000 description 2
- 108090001061 Insulin Proteins 0.000 description 2
- 108090001117 Insulin-Like Growth Factor II Proteins 0.000 description 2
- 102000048143 Insulin-Like Growth Factor II Human genes 0.000 description 2
- 102100037852 Insulin-like growth factor I Human genes 0.000 description 2
- 102100032825 Integrin alpha-8 Human genes 0.000 description 2
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 2
- 102100033493 Interleukin-3 receptor subunit alpha Human genes 0.000 description 2
- 108010002586 Interleukin-7 Proteins 0.000 description 2
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 2
- 108010028275 Leukocyte Elastase Proteins 0.000 description 2
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 2
- 102100031520 MAPK/MAK/MRK overlapping kinase Human genes 0.000 description 2
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 102100028123 Macrophage colony-stimulating factor 1 Human genes 0.000 description 2
- 102100027754 Mast/stem cell growth factor receptor Kit Human genes 0.000 description 2
- 102000018697 Membrane Proteins Human genes 0.000 description 2
- 206010027406 Mesothelioma Diseases 0.000 description 2
- 241000713869 Moloney murine leukemia virus Species 0.000 description 2
- 241000713862 Moloney murine sarcoma virus Species 0.000 description 2
- 101150064776 Msln gene Proteins 0.000 description 2
- 108010063954 Mucins Proteins 0.000 description 2
- 241000714177 Murine leukemia virus Species 0.000 description 2
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 2
- 102100022680 NKG2-D type II integral membrane protein Human genes 0.000 description 2
- 102100033174 Neutrophil elastase Human genes 0.000 description 2
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 2
- 102100040348 Peptidyl-prolyl cis-trans isomerase FKBP11 Human genes 0.000 description 2
- 102100040120 Prominin-1 Human genes 0.000 description 2
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 2
- 241000714474 Rous sarcoma virus Species 0.000 description 2
- 102100029198 SLAM family member 7 Human genes 0.000 description 2
- 102000001712 STAT5 Transcription Factor Human genes 0.000 description 2
- 108010029477 STAT5 Transcription Factor Proteins 0.000 description 2
- 102100038081 Signal transducer CD24 Human genes 0.000 description 2
- 102100037253 Solute carrier family 45 member 3 Human genes 0.000 description 2
- 108010092262 T-Cell Antigen Receptors Proteins 0.000 description 2
- 102100035268 T-cell surface protein tactile Human genes 0.000 description 2
- 108010017842 Telomerase Proteins 0.000 description 2
- 102000007000 Tenascin Human genes 0.000 description 2
- 108010008125 Tenascin Proteins 0.000 description 2
- 108010034949 Thyroglobulin Proteins 0.000 description 2
- 102000009843 Thyroglobulin Human genes 0.000 description 2
- 101800000385 Transmembrane protein Proteins 0.000 description 2
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 2
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 2
- 229960000723 ampicillin Drugs 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 239000006285 cell suspension Substances 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 229960004407 chorionic gonadotrophin Drugs 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 239000012141 concentrate Substances 0.000 description 2
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 2
- 210000000805 cytoplasm Anatomy 0.000 description 2
- 230000007547 defect Effects 0.000 description 2
- 238000007865 diluting Methods 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 210000003162 effector t lymphocyte Anatomy 0.000 description 2
- 230000002708 enhancing effect Effects 0.000 description 2
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 238000005206 flow analysis Methods 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 150000002339 glycosphingolipids Chemical class 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- 230000006801 homologous recombination Effects 0.000 description 2
- 238000002744 homologous recombination Methods 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 229940127121 immunoconjugate Drugs 0.000 description 2
- 238000011503 in vivo imaging Methods 0.000 description 2
- 229940125396 insulin Drugs 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 238000012737 microarray-based gene expression Methods 0.000 description 2
- 238000012243 multiplex automated genomic engineering Methods 0.000 description 2
- 230000001717 pathogenic effect Effects 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- 239000012071 phase Substances 0.000 description 2
- 229920001481 poly(stearyl methacrylate) Polymers 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 239000002244 precipitate Substances 0.000 description 2
- 230000009465 prokaryotic expression Effects 0.000 description 2
- KEYDJKSQFDUAGF-YIRKRNQHSA-N prostaglandin D2 ethanolamide Chemical compound CCCCC[C@H](O)\C=C\[C@@H]1[C@@H](C\C=C/CCCC(=O)NCCO)[C@@H](O)CC1=O KEYDJKSQFDUAGF-YIRKRNQHSA-N 0.000 description 2
- 108010079891 prostein Proteins 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 101150047061 tag-72 gene Proteins 0.000 description 2
- 229960002175 thyroglobulin Drugs 0.000 description 2
- 230000009258 tissue cross reactivity Effects 0.000 description 2
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 2
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- GOZMBJCYMQQACI-UHFFFAOYSA-N 6,7-dimethyl-3-[[methyl-[2-[methyl-[[1-[3-(trifluoromethyl)phenyl]indol-3-yl]methyl]amino]ethyl]amino]methyl]chromen-4-one;dihydrochloride Chemical compound Cl.Cl.C=1OC2=CC(C)=C(C)C=C2C(=O)C=1CN(C)CCN(C)CC(C1=CC=CC=C11)=CN1C1=CC=CC(C(F)(F)F)=C1 GOZMBJCYMQQACI-UHFFFAOYSA-N 0.000 description 1
- 102000005869 Activating Transcription Factors Human genes 0.000 description 1
- 108010005254 Activating Transcription Factors Proteins 0.000 description 1
- 208000010507 Adenocarcinoma of Lung Diseases 0.000 description 1
- 206010052747 Adenocarcinoma pancreas Diseases 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 102100036301 C-C chemokine receptor type 7 Human genes 0.000 description 1
- 102000009410 Chemokine receptor Human genes 0.000 description 1
- 108050000299 Chemokine receptor Proteins 0.000 description 1
- 102000019034 Chemokines Human genes 0.000 description 1
- 108010012236 Chemokines Proteins 0.000 description 1
- 108020004705 Codon Proteins 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
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 1
- 108010036395 Endoglin Proteins 0.000 description 1
- 102100037241 Endoglin Human genes 0.000 description 1
- 102100031780 Endonuclease Human genes 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000713859 FBR murine osteosarcoma virus Species 0.000 description 1
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 1
- 230000005526 G1 to G0 transition Effects 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 229920002527 Glycogen Polymers 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000716065 Homo sapiens C-C chemokine receptor type 7 Proteins 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- 101001018097 Homo sapiens L-selectin Proteins 0.000 description 1
- 101000979629 Homo sapiens Nucleoside diphosphate kinase A Proteins 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 208000029462 Immunodeficiency disease Diseases 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 1
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 1
- 102000004527 Interleukin-21 Receptors Human genes 0.000 description 1
- 108010017411 Interleukin-21 Receptors Proteins 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- 102100033467 L-selectin Human genes 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 1
- 102000012750 Membrane Glycoproteins Human genes 0.000 description 1
- 108010090054 Membrane Glycoproteins Proteins 0.000 description 1
- 241000713333 Mouse mammary tumor virus Species 0.000 description 1
- 102100023252 Nucleoside diphosphate kinase A Human genes 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 238000001190 Q-PCR Methods 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 210000000173 T-lymphoid precursor cell Anatomy 0.000 description 1
- 208000000728 Thymus Neoplasms Diseases 0.000 description 1
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 239000012491 analyte Substances 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 150000001450 anions Chemical class 0.000 description 1
- 230000003540 anti-differentiation Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 239000000611 antibody drug conjugate Substances 0.000 description 1
- 229940049595 antibody-drug conjugate Drugs 0.000 description 1
- 230000010100 anticoagulation Effects 0.000 description 1
- 239000008346 aqueous phase Substances 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 238000007664 blowing Methods 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 238000010804 cDNA synthesis Methods 0.000 description 1
- 208000035269 cancer or benign tumor Diseases 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 210000002421 cell wall Anatomy 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 238000001816 cooling Methods 0.000 description 1
- 108091008034 costimulatory receptors Proteins 0.000 description 1
- 238000005520 cutting process Methods 0.000 description 1
- 150000001945 cysteines Chemical class 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 239000003599 detergent Substances 0.000 description 1
- 239000000032 diagnostic agent Substances 0.000 description 1
- 229940039227 diagnostic agent Drugs 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 238000006073 displacement reaction Methods 0.000 description 1
- MOTZDAYCYVMXPC-UHFFFAOYSA-N dodecyl hydrogen sulfate Chemical compound CCCCCCCCCCCCOS(O)(=O)=O MOTZDAYCYVMXPC-UHFFFAOYSA-N 0.000 description 1
- 229940043264 dodecyl sulfate Drugs 0.000 description 1
- 230000007783 downstream signaling Effects 0.000 description 1
- 239000003651 drinking water Substances 0.000 description 1
- 235000020188 drinking water Nutrition 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 108010015866 endodeoxyribonuclease NheI Proteins 0.000 description 1
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 238000001976 enzyme digestion Methods 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 239000000834 fixative Substances 0.000 description 1
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 1
- 238000012632 fluorescent imaging Methods 0.000 description 1
- 238000011010 flushing procedure Methods 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 229940096919 glycogen Drugs 0.000 description 1
- 229930004094 glycosylphosphatidylinositol Natural products 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 102000027596 immune receptors Human genes 0.000 description 1
- 108091008915 immune receptors Proteins 0.000 description 1
- 230000007813 immunodeficiency Effects 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 239000012535 impurity Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000031146 intracellular signal transduction Effects 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 201000000062 kidney sarcoma Diseases 0.000 description 1
- 210000003141 lower extremity Anatomy 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 201000005249 lung adenocarcinoma Diseases 0.000 description 1
- 210000003738 lymphoid progenitor cell Anatomy 0.000 description 1
- 230000002934 lysing effect Effects 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 210000003593 megakaryocyte Anatomy 0.000 description 1
- 210000005033 mesothelial cell Anatomy 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 230000002071 myeloproliferative effect Effects 0.000 description 1
- 238000006386 neutralization reaction Methods 0.000 description 1
- 229910052759 nickel Inorganic materials 0.000 description 1
- 235000021590 normal diet Nutrition 0.000 description 1
- 210000004940 nucleus Anatomy 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 239000012044 organic layer Substances 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 201000002094 pancreatic adenocarcinoma Diseases 0.000 description 1
- 238000004091 panning Methods 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 239000000902 placebo Substances 0.000 description 1
- 229940068196 placebo Drugs 0.000 description 1
- 239000013600 plasmid vector Substances 0.000 description 1
- 231100000683 possible toxicity Toxicity 0.000 description 1
- XAEFZNCEHLXOMS-UHFFFAOYSA-M potassium benzoate Chemical compound [K+].[O-]C(=O)C1=CC=CC=C1 XAEFZNCEHLXOMS-UHFFFAOYSA-M 0.000 description 1
- 229910001414 potassium ion Inorganic materials 0.000 description 1
- 238000012257 pre-denaturation Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 238000000751 protein extraction Methods 0.000 description 1
- 238000001742 protein purification Methods 0.000 description 1
- 239000011535 reaction buffer Substances 0.000 description 1
- 108010054624 red fluorescent protein Proteins 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 239000012266 salt solution Substances 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 229910001415 sodium ion Inorganic materials 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 239000012089 stop solution Substances 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 201000009377 thymus cancer Diseases 0.000 description 1
- 210000001685 thyroid gland Anatomy 0.000 description 1
- 201000009657 thyroid sarcoma Diseases 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- 230000034512 ubiquitination Effects 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- 238000009827 uniform distribution Methods 0.000 description 1
- 239000013603 viral vector Substances 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
Classifications
-
- 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
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K19/00—Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/62—DNA sequences coding for fusion proteins
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N7/00—Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Zoology (AREA)
- General Health & Medical Sciences (AREA)
- Biomedical Technology (AREA)
- Medicinal Chemistry (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biochemistry (AREA)
- Wood Science & Technology (AREA)
- Molecular Biology (AREA)
- General Engineering & Computer Science (AREA)
- Immunology (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Cell Biology (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Biophysics (AREA)
- Virology (AREA)
- Microbiology (AREA)
- Developmental Biology & Embryology (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Epidemiology (AREA)
- Hematology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Peptides Or Proteins (AREA)
Abstract
Synthetic T cell receptor antigen receptor specifically binding mesothelin and its application are provided. The antigen binding fragments of the targeting MSLN are used for modifying TCR and CAR, so that better effect on tumor treatment is obtained.
Description
The invention relates to the field of biological medicine, in particular to a synthetic T cell receptor antigen receptor (STAR) specifically binding to mesothelin, a STAR complex, immune cells containing the STAR or STAR complex, and application thereof in the field of biological medicine.
Mesothelin (Mesothelin), also known as MSLN, is a cell surface glycoprotein encoded by the MSLN gene. The MSLN gene encodes a proprotein that is proteolytically produced to produce both megakaryocyte potentiators (megakaryocyte-potentiating factor, MPF) and mesothelin MSLN protein products. Wherein MSLN is a protein anchored to the cell surface by glycosyl phosphatidylinositol, a differentiation antigen present on normal mesothelial cells. MSLN is rarely expressed in normal tissues, but MSLN is found to be overexpressed in a variety of cancers, and thus mesothelin MSLN is likely to be an important target for cancer treatment. The overexpression of mesothelin is currently observed in mesothelioma, ovarian cancer, lung cancer, esophageal cancer, pancreatic cancer, gastric cancer, cholangiocarcinoma, endometrial cancer, thymus cancer, colon cancer and breast cancer. The frequency and distribution of MSLN expression varies with tumor subtype. In cancer cells, MSLN expression may be in the lumen/membrane or cytoplasm. There is also a certain difference in the expression position of different types of tumor MSLNs. In mesothelioma tumors, MSLN is expressed on the cell surface in a uniform distribution. In lung adenocarcinoma, MSLN is expressed both on the cytoplasm and on the cell surface. In gastric cancer, cytoplasmic expression is more prevalent than membrane expression. MSLN is also expressed in solid tumors such as thyroid, renal and synovial sarcoma tumors.
For tumor treatment, CAR-T and TCR-T cells currently develop rapidly, but STAR-T derived from the native TCR lacks co-stimulatory signals in T cell activation during activation, and its proliferation and activation capacity is often affected. Thus, there remains a need in the art for improved TCRs and corresponding TCR-T therapies.
Therefore, the application modifies TCR and CAR with antigen binding fragments targeting MSLN, and is expected to obtain better effect on tumor treatment.
Disclosure of Invention
In order to solve the defects in the prior art, the invention provides a synthetic T cell receptor antigen receptor STAR and application thereof, wherein the synthetic T cell receptor antigen receptor can specifically bind mesothelin, and the TCR and the CAR are modified through an antigen binding fragment of targeting MSLN, so that a better tumor treatment effect is obtained.
In particular, in a first aspect of the invention, there is provided a synthetic T cell receptor antigen receptor (STAR),
I) The synthetic T cell receptor antigen receptor comprises an alpha chain comprising a first target binding region and a first constant region, and a beta chain comprising a second target binding region and a second constant region.
Or alternatively
Ii) the synthetic T cell receptor antigen receptor comprises a gamma chain, a delta chain, wherein the gamma chain comprises a first target binding region and a first constant region, and the delta chain comprises a second target binding region and a second constant region. Preferably, i) the alpha chain and/or the beta chain has attached at its C-terminal end at least one functional domain; or ii) the gamma and/or delta chain has at least one functional domain attached to its C-terminus.
It is further preferred that i) said at least one functional domain is linked directly or via a linker to the C-terminus of the alpha and/or beta chain, or ii) said at least one functional domain is linked directly or via a linker to the C-terminus of the gamma and/or delta chain.
Preferably, i) the intracellular regions of the alpha and/or beta chain in the synthetic T cell receptor antigen receptor are deleted; or ii) the intracellular region of the gamma and/or delta chain in the synthetic T cell receptor antigen receptor is deleted.
Still further preferred, the functional domain of i) is linked directly or via a linker to the C-terminus of the alpha and/or beta chain deleted from the intracellular region; or ii) the functional domain is linked directly or via a linker to the C-terminus of the gamma and/or delta chain deleted from the intracellular region.
Preferably, i) the C-terminal linkage of the alpha chain in the synthetic T cell receptor antigen receptor is 1,2, 3, 4, 5,6,7,8, 9, 10 or
More functional domains, and/or, 1,2, 3, 4, 5, 6, 7, 8, 9, 10 or more functional domains linked at the C-terminus of the β -chain in said synthetic T-cell receptor antigen receptor; or ii) 1,2, 3, 4, 5, 6, 7, 8, 9, 10 or more functional domains are C-terminally linked to the gamma chain in the synthetic T cell receptor antigen receptor, and/or 1,2, 3, 4, 5, 6, 7, 8, 9, 10 or more functional domains are C-terminally linked to the delta chain in the STAR.
In one embodiment of the invention, the multiple (including two or more) functional domains are linked directly or through a linker.
In one embodiment of the invention, the synthetic T cell receptor antigen receptor has at least one functional domain attached to the C-terminus of the alpha chain. The intracellular region of the alpha chain is deleted. The functional domain is linked to the C-terminus of the alpha chain deleted in the intracellular region by a linker.
In one embodiment of the invention, the synthetic T cell receptor antigen receptor has at least one functional domain attached to the C-terminus of the β -chain. The intracellular region of the beta chain is deleted. The functional domain is linked to the C-terminus of the beta chain deleted in the intracellular region by a linker.
In one embodiment of the invention, the synthetic T cell receptor antigen receptor has at least one functional domain attached to the C-terminus of the gamma chain. The intracellular region of the gamma chain is deleted. The functional domain is linked to the C-terminus of the gamma chain deleted in the intracellular region by a linker.
In one embodiment of the invention, the delta chain of the synthetic T cell receptor antigen receptor is linked at the C-terminus to at least one functional domain. The intracellular region of the delta chain is deleted. The functional domain is linked to the C-terminus of the delta chain deleted in the intracellular region by a linker.
Preferably, i) the functional domains of the C-terminal linkage of the alpha and/or beta chains in the synthetic T cell receptor antigen receptor are the same or different;
Or ii) the functional domains of the gamma chain and/or delta chain C-terminal linkage in the synthetic T cell receptor antigen receptor are the same or different.
In one embodiment of the invention, the multiple functional domains of the synthetic T cell receptor antigen receptor to which the alpha chain is linked may be the same or different.
In one embodiment of the invention, the plurality of functional domains of the β -chain linkage in the synthetic T-cell receptor antigen receptor may be the same or different.
In one embodiment of the invention, the plurality of functional domains linked by the gamma chain in the synthetic T cell receptor antigen receptor may be the same or different.
In one embodiment of the invention, the multiple functional domains linked by the delta chain in the synthetic T cell receptor antigen receptor may be the same or different.
In one embodiment of the invention, the functional domain of the alpha chain linkage and the functional domain of the beta chain linkage in the synthetic T cell receptor antigen receptor may be the same or different.
In one embodiment of the invention, the functional domain of the gamma chain linkage and the functional domain of the delta chain linkage in the synthetic T cell receptor antigen receptor may be the same or different.
Preferably, the functional domain is a co-stimulatory molecule or a fragment thereof, a co-inhibitory molecule or a fragment thereof, a cytokine receptor or a fragment thereof, or an intracellular protein or a fragment thereof. Further preferred, the functional domain is an intracellular domain of a co-stimulatory molecule, an intracellular domain of a co-inhibitory molecule, an intracellular domain of a cytokine receptor or an intracellular protein. It may also be a fusion of the intracellular domain of the cytokine receptor with a human STAT5 activation module (amino acid sequence shown in SEQ ID NO: 25), either directly or via a linker.
The co-stimulatory molecule is selected from the group consisting of CD40, OX40, ICOS, CD28, 4-1BB (CD 137) or CD27.
The co-inhibitory molecules are selected from TIM3, PD1, CTLA4 and LAG3.
The cytokine receptor is selected from interleukin receptor (such as IL-2 receptor), interferon receptor, tumor necrosis factor superfamily receptor, colony stimulating factor receptor, chemokine receptor, growth factor receptor or other membrane proteins.
The intracellular protein is a T cell regulator, such as the domain of NIK.
In one embodiment of the invention, the co-stimulatory molecule is CD40, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:10, and a polypeptide having the amino acid sequence shown in FIG. 10.
In one embodiment of the invention, the co-stimulatory molecule is OX40, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:11, and a polypeptide comprising the amino acid sequence shown in seq id no.
In one embodiment of the invention, the co-stimulatory molecule is ICOS, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:12, and a polypeptide having the amino acid sequence shown in FIG. 12.
In one embodiment of the invention, the co-stimulatory molecule is CD28, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:13, and a nucleotide sequence shown in seq id no.
In one embodiment of the invention, the co-stimulatory molecule is 4-1BB, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:14, and a polypeptide having the amino acid sequence shown in seq id no.
In one embodiment of the invention, the co-stimulatory molecule is CD27, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:15, and a polypeptide having the amino acid sequence shown in seq id no.
In one embodiment of the invention, the cytokine receptor is IL-2β, the intracellular domain of which comprises the sequence set forth in SEQ ID NO:22, and a polypeptide comprising the amino acid sequence shown in seq id no.
In one embodiment of the invention, the cytokine receptor is IL-7α, the intracellular domain of which comprises the sequence set forth in SEQ ID NO: 23.
In one embodiment of the invention, the cytokine receptor is IL-21, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:24, and a nucleotide sequence shown in seq id no.
In a specific embodiment of the invention, the functional domain is a fusion of the IL-2β intracellular domain with a human STAT5 activating module comprising the amino acid sequence of SEQ ID NO:26, and a polypeptide comprising the amino acid sequence shown in seq id no.
In a specific embodiment of the invention, the functional domain is a fusion of the IL-7α intracellular domain with a human STAT5 activation module comprising the sequence of SEQ ID NO: 27.
Preferably, the first constant region is a TCR α chain constant region or a TCR γ chain constant region, preferably a modified TCR α chain constant region or a modified TCR γ chain constant region.
The TCR a chain constant region is selected from a human TCR a chain constant region or a rodent (preferably murine, further preferably mouse) TCR a chain constant region. The TCR gamma chain constant region is selected from a human TCR gamma chain constant region or a rodent (preferably murine, further preferably mouse) TCR gamma chain constant region.
In one embodiment of the invention, the amino acid sequence of the human TCR α chain constant region is as set forth in SEQ ID NO:1, the amino acid sequence of the constant region of the TCR alpha chain of said rodent (preferably murine, more preferably mouse) is as set forth in SEQ ID NO: 3.
In one embodiment of the present invention, the amino acid sequence of the human TCR gamma chain constant region is as set forth in SEQ ID NO:45, the amino acid sequence of the constant region of the murine (preferably murine, more preferably mouse) TCR gamma chain is as set forth in SEQ ID NO: 46.
The modified TCR a chain constant region is derived from a human TCR a chain constant region comprising one or more modifications at positions 48, 116 or 119 relative to a wild-type human TCR a chain constant region, the modifications being mutations or deletions.
The modified TCR a chain constant region is derived from a human TCR a chain constant region comprising a threonine T mutation at position 48 to cysteine C relative to a wild-type human TCR a chain constant region.
The modified TCR alpha chain constant region is derived from a human TCR alpha chain constant region which is mutated to leucine L at serine S116 and valine V at glycine G119 relative to a wild-type human TCR alpha chain constant region.
The modified TCR alpha chain constant region is derived from a human TCR alpha chain constant region comprising a mutation of threonine T to cysteine C at position 48, serine S to leucine L at position 116, and glycine G to valine V at position 119, relative to a wild-type human TCR alpha chain constant region.
The modified TCR a chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region comprising one or more modifications at positions 6, 13, 15-18, 48, 112, 114, 115, relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region, the modifications being mutations or deletions.
The modified TCR a chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region comprising one or more modifications at positions 13, 36, 47, 53, 58, 78, 98, 122, relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region, the modifications being mutations or deletions.
Preferably, the modified TCR a chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region comprising the introduction of cysteine relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region.
Further preferred, the modified TCR a chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR a chain constant region comprising an amino acid, e.g. threonine T, at position 48 mutated to cysteine C relative to a wild-type rodent (preferably murine, further preferred mouse) TCR a chain constant region.
In a specific embodiment of the invention, a rodent (preferably a mouse, more preferably a mouse) TCR a chain constant region comprising a cysteine introduction comprises the amino acid sequence of SEQ ID NO: 5.
Preferably, the modified TCR a chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region comprising a hydrophobic amino acid mutation relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region.
Further preferred, the modified TCR α chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR α chain constant region comprising an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, and/or an amino acid such as glycine G at position 115 changed to valine V, relative to a wild-type rodent (preferably murine, further preferred mouse) TCR α chain constant region.
In a specific embodiment of the invention, a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising a hydrophobic amino acid mutation comprises the amino acid sequence of SEQ ID NO: 7.
Preferably, the modified TCR a chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region comprising an N-terminal modification relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region.
Further preferred, the modified TCR α chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR α chain constant region comprising an amino acid at position 6, e.g. E, substituted with D, K at position 13, substituted with R, and amino acids 15-18 deleted relative to a wild-type rodent (preferably murine, further preferred mouse) TCR α chain constant region.
Preferably, the modified TCR a chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising a lysine to arginine mutation of the transmembrane region.
Further preferred, the modified TCR α chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR α chain constant region comprising the amino acid K at position 122 substituted with R relative to a wild-type rodent (preferably murine, further preferred mouse) TCR α chain constant region.
In a specific embodiment of the invention, a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising a transmembrane region lysine mutated to arginine comprises SEQ ID NO: 8.
Preferably, the modified TCR a chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region comprising a cysteine introduction and a hydrophobic amino acid mutation relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region.
Further preferred, the modified TCR α chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR α chain constant region comprising an amino acid such as threonine T at position 48 mutated to cysteine C, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V, relative to a wild-type rodent (preferably murine, further preferred mouse) TCR α chain constant region.
In a specific embodiment of the invention, a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising a cysteine introduction and a hydrophobic amino acid mutation comprises the amino acid sequence of SEQ ID NO: 31.
The modified TCR a chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising an amino acid at position 48, e.g. threonine T, mutated to cysteine C, with amino acid K at position 122 being substituted with R, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR a chain constant region.
The modified TCR a chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, and an amino acid such as threonine T at position 48 mutated to cysteine C, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR a chain constant region.
The modified TCR α chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR α chain constant region comprising an amino acid such as threonine T mutated to cysteine C at position 48, an amino acid such as serine S changed to leucine L at position 112, an amino acid such as methionine M changed to isoleucine I at position 114, an amino acid such as glycine G changed to valine V at position 115, and an amino acid K replaced by R, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR α chain constant region.
The modified TCR α chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR α chain constant region comprising an amino acid such as E substituted at position 6 with D, K substituted at position 13 with R, and amino acids 15-18 deleted, an amino acid such as threonine T mutated to cysteine C, an amino acid such as serine S at position 112 with leucine L, an amino acid such as methionine M at position 114 with isoleucine I, an amino acid such as glycine G at position 115 with valine V, an amino acid K substituted at position 122 with R, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR α chain constant region.
The modified TCR α chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, an amino acid such as threonine T at position 48 mutated to cysteine C, amino acid K at position 122 substituted with R, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR α chain constant region.
The modified TCR α chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR α chain constant region.
The modified TCR α chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, and amino acid K at position 122 substituted with R, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR α chain constant region.
The modified TCR α chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V, and an amino acid K at position 122 replaced with R, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR α chain constant region.
Preferably, the modified TCR a chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region comprising a cysteine introduction, a hydrophobic amino acid mutation and an N-terminal modification relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR a chain constant region.
Further preferred, the modified TCR α chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, amino acids 15-18 deleted, amino acid such as threonine T mutated to cysteine C at position 48, amino acid such as serine S to leucine L, amino acid such as methionine M at position 114 to isoleucine I, amino acid such as glycine G at position 115 to valine V, relative to a wild-type rodent (preferably murine, further preferred mouse) TCR α chain constant region.
In a specific embodiment of the invention, a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising a cysteine introduction, a hydrophobic amino acid mutation, and a deletion of an intracellular region comprises the amino acid sequence of SEQ ID NO:16, and a polypeptide having the amino acid sequence shown in 16.
Preferably, the modified TCR a chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising an N-terminal modification, a cysteine introduction and a hydrophobic amino acid mutation.
Further preferred, the modified TCR α chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR α chain constant region comprising an amino acid such as E substituted with D, K substituted with R at position 13, amino acids 15-18 deleted, and amino acid such as threonine T mutated to cysteine C at position 48, amino acid such as serine S changed to leucine L at position 112, amino acid such as methionine M changed to isoleucine I at position 114, amino acid such as glycine G changed to valine V at position 115.
In a specific embodiment of the invention, a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising an N-terminal modification, a cysteine introduction and a hydrophobic amino acid mutation comprises SEQ ID NO:32, and a polypeptide having the amino acid sequence shown in seq id no.
Preferably, the modified TCR a chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising an intracellular deletion, an N-terminal modification, a cysteine introduction and a hydrophobic amino acid mutation.
Further preferred, the modified TCR α chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR α chain constant region comprising an intracellular deletion, and the amino acid, e.g. threonine T, at position 48 is mutated to cysteine C, the amino acid, e.g. serine S, at position 112 is changed to leucine L, the amino acid, e.g. methionine M, at position 114 is changed to isoleucine I, the amino acid, e.g. glycine G, at position 115 is changed to valine V, and the amino acid, e.g. E, at position 6 is substituted with D, K at position 13 is substituted with R, and the amino acids at positions 15-18 are deleted.
In a specific embodiment of the invention, a rodent (preferably murine, further preferably mouse) TCR a chain constant region comprising an intracellular deletion, an N-terminal modification, a cysteine introduction and a hydrophobic amino acid mutation comprises the amino acid sequence of SEQ ID NO: 43.
In a specific embodiment of the invention, the first constant region comprises SEQ ID NO: 1.3, 5, 7, 8, 16, 31, 32 or 43.
Preferably, the second constant region is a TCR β chain constant region or a TCR δ chain constant region, preferably a modified TCR β chain constant region or a modified TCR δ chain constant region.
Further preferred, the TCR β chain constant region is selected from a human TCR β chain constant region or a rodent (preferably murine, further preferred mouse) TCR β chain constant region, and the TCR δ chain constant region is selected from a human TCR δ chain constant region or a rodent (preferably murine, further preferred mouse) TCR δ chain constant region.
In one embodiment of the invention, the human TCR β chain constant region comprises SEQ ID NO:2, and a polypeptide having the amino acid sequence shown in 2.
In a specific embodiment of the invention, the rodent (preferably murine, further preferably mouse) TCR β chain constant region comprises the amino acid sequence of SEQ ID NO:4, and a polypeptide having the amino acid sequence shown in (a) and (b).
In one embodiment of the invention, the human TCR delta chain constant region comprises SEQ ID NO: 47.
In a specific embodiment of the invention, the rodent (preferably murine, further preferably mouse) TCR delta chain constant region comprises the amino acid sequence of SEQ ID NO: 48.
The modified TCR β chain constant region is derived from a human TCR β chain constant region comprising one or more modifications at position 57, 173, or 175 relative to a wild-type human TCR β chain constant region, the modifications being mutations or deletions.
The modified TCR β chain constant region is derived from a human TCR β chain constant region comprising a serine S mutation at position 57 to cysteine C relative to a wild-type human TCR β chain constant region.
The modified TCR β chain constant region is derived from a human TCR β chain constant region comprising lysine K mutations to arginine at positions 173 and 175 relative to a wild-type human TCR β chain constant region.
The modified TCR β chain constant region is derived from a human TCR β chain constant region comprising a serine S mutation at position 57 to cysteine C, and lysine K mutations at positions 173 and 175 to arginine, relative to a wild-type human TCR β chain constant region.
The modified TCR β chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region comprising one or more modifications at positions 3, 6, 9, 11, 12, 17, 21-25, 56, 150, 168 or 170 relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region, the modifications being mutations or deletions.
The modified TCR β chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region comprising one or more modifications at positions 9, 17, 23, 25, 49, 63, 103, 110, 150, 168, 170 relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region, the modifications being mutations or deletions.
Preferably, the modified TCR β chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region comprising the introduction of cysteine relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region.
Further preferred, the modified TCR β chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR β chain constant region, which is mutated to cysteine C at amino acid 56, e.g. serine S, relative to a wild-type rodent (preferably murine, further preferred mouse) TCR β chain constant region.
In a specific embodiment of the invention, the cysteine-containing introduced rodent (preferably murine, further preferably mouse) TCR β chain constant region comprises the amino acid sequence of SEQ ID NO:6, and a polypeptide having the amino acid sequence shown in FIG. 6.
Preferably, the modified TCR β chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR β chain constant region, the intracellular region lysine of which is replaced by arginine.
Further preferred, the modified TCR β chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR β chain constant region, the lysine at position 150, 168 or 170 of which is substituted with arginine.
In a specific embodiment of the invention, a rodent (preferably murine, further preferably mouse) TCR β chain constant region comprising an intracellular region lysine replaced with arginine comprises SEQ ID NO:9, and a nucleotide sequence shown in FIG. 9
Preferably, the modified TCR β chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region comprising an N-terminal modification relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region.
Further preferred, the modified TCR β chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR β chain constant region which is substituted with K for amino acids such as R at position 3, F for amino acids such as T at position 6, E for K at position 9, a for S at position 11, V for L at position 12, and deleted at amino acids 17, 21-25 relative to a wild-type rodent (preferably murine, further preferred mouse) TCR β chain constant region.
Preferably, the modified TCR β chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region comprising the introduction of cysteines and the deletion of an intracellular region relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region.
In a specific embodiment of the invention, the rodent (preferably murine, further preferably mouse) TCR β chain constant region comprising the introduction of a cysteine and the deletion of an intracellular region comprises the sequence set forth in SEQ ID NO:17, and a sequence of amino acids shown in seq id no.
The modified TCR β chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR β chain constant region which comprises an amino acid at position 56, such as serine S, mutated to cysteine C, and a lysine at position 150, 168 or 170 substituted with arginine relative to a wild-type rodent (preferably murine, further preferably mouse) TCR β chain constant region.
The modified TCR β chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR β chain constant region comprising an amino acid such as R substituted with K, an amino acid such as T substituted with F, K substituted with E, S substituted with a, L substituted with V, and amino acids 17, 21-25 deleted, an amino acid such as serine S mutated to cysteine C, at position 56, and lysine substituted with arginine at position 150, 168 or 170, at position 6, such as T substituted with F, K substituted with E, S substituted with S at position 11, V substituted with L at position 12.
The modified TCR β chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR β chain constant region comprising an amino acid such as R substituted with K, an amino acid such as T substituted with F, K substituted with E, S substituted with a, L substituted with V, and amino acids 17, 21-25 deleted, and lysine substituted with arginine at positions 150, 168 or 170 at position 6, such as T, at position 9, at position 11, relative to a wild-type rodent (preferably murine, further preferably mouse).
Preferably, the modified TCR β chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region comprising an N-terminal modification and a cysteine introduction relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region.
The modified TCR β chain constant region is derived from a rodent (preferably murine, further preferably mouse) TCR β chain constant region comprising an amino acid such as R at position 3 substituted with K, an amino acid such as T at position 6 substituted with F, K at position 9 substituted with E, S at position 11 substituted with a, L at position 12 substituted with V, and amino acids 17, 21-25 deleted, and an amino acid such as serine S at position 56 mutated to cysteine C, relative to a wild-type rodent (preferably murine, further preferably mouse) TCR β chain constant region.
In a specific embodiment of the invention, the rodent (preferably murine, further preferably mouse) TCR β chain constant region comprising an N-terminal modification and a cysteine introduction comprises SEQ ID NO:33, and a nucleotide sequence shown in seq id no.
Preferably, the modified TCR β chain constant region is derived from a rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region comprising an intracellular deletion, an N-terminal modification and a cysteine introduction relative to a wild-type rodent (preferably a mouse, further preferably a mouse) TCR β chain constant region.
Further preferred, the modified TCR β chain constant region is derived from a rodent (preferably murine, further preferred mouse) TCR β chain constant region comprising an intracellular deletion relative to a wild-type rodent (preferably murine, further preferred mouse) TCR β chain constant region, and the amino acid at position 56, e.g. serine S, is mutated to cysteine C, and the amino acid at position 3, e.g. R, is substituted with K, the amino acid at position 6, e.g. T, is substituted with F, the K at position 9 is substituted with E, the S at position 11 is substituted with a, the L at position 12 is substituted with V, and the amino acids at positions 17, 21-25 are deleted.
In a specific embodiment of the invention, the rodent (preferably murine, further preferably mouse) TCR β chain constant region comprising an intracellular deletion, an N-terminal modification and a cysteine introduction comprises SEQ ID NO:44, and a polypeptide having the amino acid sequence shown in seq id no.
In a specific embodiment of the invention, the second constant region comprises SEQ ID NO: 2.4, 6, 9, 17, 33 or 44.
The target binding region is located at the N-terminus of the constant region. The two may be connected directly or through a joint.
The first target binding zone may comprise one or more identical or different binding zones. The target binding region is an antigen binding region or fragment thereof, a non-immunoglobulin antigen binding domain or fragment thereof, an antibody binding region or fragment thereof, a receptor or fragment thereof, a ligand or fragment thereof, preferably a natural T cell receptor.
The antigen binding region is derived from an antibody.
The STAR comprises one or more antigen binding regions;
preferably, the plurality of antigen binding regions are the same or different;
Further preferred, the plurality of antigen binding regions are linked directly or through a linker.
The antibody may be a monoclonal antibody or a polyclonal antibody.
The antibody may also comprise a fragment such as F ab、F ab'、F ab'-SH、Fv、scFv、(F ab') 2, a single domain antibody, a diabody (dAb) or a linear antibody.
The antibody may be a monospecific antibody or a multispecific antibody (e.g., bispecific antibody).
Preferably, the antibody may be a fully human antibody, a humanized antibody, an animal-derived antibody. Wherein the animal can be mouse, rabbit, cow, monkey, etc.
Preferably, the first antigen binding region is linked directly to the first constant region or via a linker and/or the second antigen binding region is linked directly to the second constant region or via a linker.
Preferably, the first antigen binding region and the second antigen binding region each, independently or in combination, specifically bind to a target antigen.
Preferably, the target antigen is a disease-associated antigen, preferably a cancer-associated antigen, for example a cancer-associated antigen selected from the group consisting of: GPC3, CD16, CD64, CD78, CD96, CLL1, CD116, CD117, CD71, CD45, CD71, CD123, CD138, erbB2 (HER 2/neu), carcinoembryonic antigen (CEA), epithelial cell adhesion molecule (EpCAM), epidermal Growth Factor Receptor (EGFR), EGFR variant III (EGFRvIII), CD19, CD20, CD30, CD40, bissialoglycganglioside GD2, ductal mucin, gp36, TAG-72, glycosphingolipids, glioma-associated antigen, beta-human chorionic gonadotrophin, alpha Fetoprotein (AFP), lectin-reactive AFP, thyroglobulin, RAGE-1, MN-CA IX, human telomerase reverse transcriptase, RU1, RU2 (AS), enterocarboxylesterase, mut hsp70-2, M-CSF prostase (prostase), prostase Specific Antigen (PSA), PAP, NY-ESO-1, LAGA-1a, p53, prostein, PSMA, survival and telomerase, prostate cancer tumor antigen-1 (PCTA-1), MAGE, ELF2M, neutrophil elastase, ephrin B2, CD22, insulin growth factor (IGF 1) -I, IGF-II, IGFI receptor, mesothelin, major Histocompatibility Complex (MHC) molecules presenting tumor specific peptide epitopes, 5T4, ROR1, nkp, NKG2D, tumor matrix antigen, additional domain a (EDA) and additional domain B (EDB) of fibronectin, A1 domain of tenascin-C (TnC A1), fibroblast-related protein (fap), CD3, CD4, CD8, CD24, fibronectin, CD25, CD33, CD34, CD133, CD138, foxp3, B7-1 (CD 80), B7-2 (CD 86), GM-CSF, cytokine receptor, endoglin, major Histocompatibility Complex (MHC) molecule 、BCMA(CD269、TNFRSF17)、TNFRSF17(UNIPROTQ02223)、SLAMF7(UNIPROT Q9NQ25)、GPRC5D(UNIPROT Q9NZD1)、FKBP11(UNIPROT Q9NYL4)、KAMP3、ITGA8(UNIPROT P53708), or FCRL5 (UNPROT Q68SN 8).
Preferably, the first antigen binding region comprises the heavy chain variable region of one or more antibodies that specifically bind the target antigen, and the second antigen binding region comprises the light chain variable region of one or more antibodies that specifically bind the target antigen; or the first antigen binding region comprises the light chain variable region of one or more antibodies that specifically bind to the target antigen and the second antigen binding region comprises the heavy chain variable region of one or more antibodies that specifically bind to the target antigen.
Wherein the heavy chain variable regions of the plurality of antibodies are linked directly or through a linker. The light chain variable regions of the plurality of antibodies are linked directly or through a linker.
Preferably, the first antigen binding region comprises one or more single chain antibodies or one or more single domain antibodies that specifically bind to the target antigen; and/or the second antigen binding region comprises one or more single chain antibodies or one or more single domain antibodies that specifically bind to the target antigen.
Wherein the plurality of single chain antibodies are directly linked or linked by a linker. The plurality of single domain antibodies are linked directly or through a linker.
Preferably, the single chain antibody comprises a heavy chain variable region and a light chain variable region linked directly or via a linker.
Preferably, the first antigen binding region and the second antigen binding region bind the same or different target antigens.
Preferably, the first antigen binding region and the second antigen binding region bind to different regions (e.g., different epitopes) of the same target antigen.
In one embodiment of the invention, the target antigen is mesothelin.
In one embodiment of the invention, the first antigen binding region comprises one or more single domain antibodies and/or the second antigen binding region comprises one or more single domain antibodies;
Preferably, the plurality of single domain antibodies comprised in the first antigen binding region are the same or different;
Preferably, the second antigen binding region comprises a plurality of single domain antibodies that are the same or different;
Further preferred, the plurality of single domain antibodies are linked directly or through a linker.
Preferably, the first antigen binding region comprises a single domain antibody that is the same or different from the single domain antibody comprising the second antigen binding region.
In one embodiment of the invention, the single domain antibody comprises a heavy chain variable region comprising CDR1-3, wherein,
I) CDR1 comprises SEQ ID NO:34, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:35, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:36, and a nucleotide sequence shown in seq id no;
Or alternatively
Ii) CDR1 comprises SEQ ID NO:37, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:38, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:39, and a polypeptide having the amino acid sequence shown in seq id no.
In one embodiment of the invention, the single domain antibody comprises SEQ ID NO:28 or 29.
In a second aspect of the present invention, there is provided a STAR complex, wherein,
I) The STAR complex comprises an alpha chain comprising a first target binding region and a first constant region, a beta chain comprising a second target binding region and a second constant region, CD3 epsilon, CD3 gamma, CD3 delta, and CD3 zeta;
Or alternatively
Ii) the STAR complex comprises a gamma chain comprising a first target binding region and a first constant region, a delta chain comprising a second target binding region and a second constant region, CD3 epsilon, CD3 gamma, CD3 delta, and CD3 zeta.
Preferably, i) at least one of the α chain, β chain, cd3ε, cd3γ, cd3δ and cd3ζ has at least one functional domain attached at its C-terminus; or ii) at least one of the gamma chain, delta chain, CD3 epsilon, CD3 gamma, CD3 delta and CD3 zeta has at least one functional domain attached to its C-terminus.
Preferably, i) said at least one functional domain is linked directly or through a linker to the C-terminus of at least one of the α chain, β chain, CD3 epsilon, CD3 gamma, CD3 delta and CD3 zeta; or ii) said at least one functional domain is linked directly or through a linker to the C-terminus of at least one of the gamma chain, delta chain, CD3 epsilon, CD3 gamma, CD3 delta and CD3 zeta.
Preferably, the intracellular region of at least one of the α chain, β chain, cd3ε, cd3γ, cd3δ, and cd3ζ in said STAR complex is deleted; or ii) the intracellular region of at least one of the gamma chain, delta chain, CD3 epsilon, CD3 gamma, CD3 delta, and CD3 zeta in said STAR complex.
Further preferred, i) said at least one functional domain is linked directly or via a linker to the C-terminus of at least one of the alpha chain, beta chain, CD3 epsilon, CD3 gamma, CD3 delta and CD3 zeta deleted of the intracellular region; or ii) said at least one functional domain is linked directly or through a linker to the C-terminus of at least one of the gamma chain, delta chain, CD3 epsilon, CD3 gamma, CD3 delta, and CD3 zeta deleted of the intracellular region.
In a specific embodiment of the invention, i) the C-terminal of at least one of the α chain, β chain, CD3 epsilon, CD3 gamma, CD3 delta, and CD3 zeta in the STAR complex is linked to 1,2, 3, 4, 5, 6, 7, 8, 9, 10 or more functional domains; or ii) at least one of the C-terminal linkages 1,2, 3, 4, 5, 6, 7, 8, 9, 10 or more functional domains of at least one of the gamma chain, delta chain, CD3 epsilon, CD3 gamma, CD3 delta, and CD3 zeta in the STAR complex.
Preferably, the multiple functional domains can be directly connected or connected through a linker.
Preferably, the C-terminal linked functional domains of at least one of the α chain, β chain, cd3ε, cd3γ, cd3δ, and cd3ζ in said STAR complex are the same or different; or ii) the C-terminal linked functional domains of at least one of the gamma chain, delta chain, CD3 epsilon, CD3 gamma, CD3 delta, and CD3 zeta in said STAR complex are the same or different.
In one embodiment of the invention, the multiple functional domains of the alpha chain linkage may be the same or different. The multiple functional domains of the beta chain linkage may be the same or different. The multiple functional domains of the gamma chain linkage may be the same or different. The multiple functional domains of the delta chain linkage may be the same or different. The multiple functional domains of the CD3 epsilon linkage may be the same or different. The multiple functional domains of the cd3γ linkage may be the same or different. The multiple functional domains of the CD3 delta linkage may be the same or different. The multiple functional domains of the cd3ζ connection may be the same or different.
In one embodiment of the invention, the functional domains to which the α chain, β chain, cd3ε, cd3γ, cd3δ and cd3ζ are linked, respectively, may be the same or different.
In one embodiment of the invention, the functional domains to which the gamma chain, delta chain, CD3 epsilon, CD3 gamma, CD3 delta and CD3 zeta chains are linked, respectively, may be the same or different.
Preferably, the functional domain is a co-stimulatory molecule or a fragment thereof, a co-inhibitory molecule or a fragment thereof, a cytokine receptor or a fragment thereof, or an intracellular protein or a fragment thereof. Further preferred, the functional domain is an intracellular domain of a co-stimulatory molecule, an intracellular domain of a co-inhibitory molecule, an intracellular domain of a cytokine receptor or an intracellular protein;
Preferably, the co-stimulatory molecule is selected from the group consisting of CD40, OX40, ICOS, CD28, 4-1BB (CD 137) or CD27;
preferably, the co-inhibitory molecule is selected from the group consisting of TIM3, PD1, CTLA4, LAG3;
Preferably, the cytokine receptor is selected from the group consisting of interleukin receptor (e.g., IL-2 receptor), interferon receptor, tumor necrosis factor superfamily receptor, colony stimulating factor receptor, chemokine receptor, growth factor receptor, and other membrane proteins;
preferably, the intracellular protein is a domain of a T cell regulator, such as NIK.
In one embodiment of the invention, the co-stimulatory molecule is CD40, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:10, and a polypeptide having the amino acid sequence shown in FIG. 10.
In one embodiment of the invention, the co-stimulatory molecule is OX40, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:11, and a polypeptide comprising the amino acid sequence shown in seq id no.
In one embodiment of the invention, the co-stimulatory molecule is ICOS, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:12, and a polypeptide having the amino acid sequence shown in FIG. 12.
In one embodiment of the invention, the co-stimulatory molecule is CD28, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:13, and a nucleotide sequence shown in seq id no.
In one embodiment of the invention, the co-stimulatory molecule is 4-1BB, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:14, and a polypeptide having the amino acid sequence shown in seq id no.
In one embodiment of the invention, the co-stimulatory molecule is CD27, the intracellular domain of which comprises the amino acid sequence of SEQ ID NO:15, and a polypeptide having the amino acid sequence shown in seq id no.
In one embodiment of the present invention, the STAR complex comprises the STAR described above, as well as CD3 ε, CD3 γ, CD3 δ, and CD3 ζ.
Preferably, said CD3 epsilon, CD3 gamma, CD3 delta and/or CD3 zeta are of human origin.
In a specific embodiment of the present invention, said CD3 epsilon comprises the amino acid sequence of SEQ ID NO:20, and a polypeptide having the amino acid sequence shown in seq id no.
In a specific embodiment of the invention, the cd3γ comprises the amino acid sequence of SEQ ID NO:18, and a polypeptide having the amino acid sequence shown in seq id no.
In a specific embodiment of the invention, the cd3δ comprises the amino acid sequence of SEQ ID NO:19, and a polypeptide comprising the amino acid sequence shown in seq id no.
In a specific embodiment of the invention, said cd3ζ comprises the amino acid sequence of SEQ ID NO:21, and a polypeptide comprising the amino acid sequence shown in seq id no.
In a third aspect of the invention, there is provided an antibody or antigen binding fragment comprising a heavy chain variable region and/or a light chain variable region.
The heavy chain variable region comprises CDR1-3, wherein,
I) CDR1 comprises SEQ ID NO:34, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:35, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:36, and a nucleotide sequence shown in seq id no.
Or alternatively
Ii) CDR1 comprises SEQ ID NO:37, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:38, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:39, and a polypeptide having the amino acid sequence shown in seq id no.
The antibody may also comprise a fragment such as F ab、F ab'、F ab'-SH、Fv、scFv、(F ab') 2, a single domain antibody, a diabody (dAb) or a linear antibody.
The antibody may be a monospecific antibody or a multispecific antibody (e.g., bispecific antibody).
Preferably, the antibody may be a fully human antibody, a humanized antibody, an animal-derived antibody. Wherein the animal can be mouse, rabbit, cow, monkey, etc.
In one embodiment of the invention, the antibody or antigen binding fragment is a single chain antibody or a single domain antibody.
In one embodiment of the invention, the antibody or antigen binding fragment comprises SEQ ID NO:28 or 29.
In a fourth aspect of the invention, there is provided a single chain antibody comprising a heavy chain variable region and/or a light chain variable region.
The heavy chain variable region comprises CDR1-3, wherein,
I) CDR1 comprises SEQ ID NO:34, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:35, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:36, and a nucleotide sequence shown in seq id no.
Or alternatively
Ii) CDR1 comprises SEQ ID NO:37, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:38, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:39, and a polypeptide having the amino acid sequence shown in seq id no.
In one embodiment of the invention, the antibody or antigen binding fragment comprises SEQ ID NO:28 or 29.
In a fifth aspect of the invention, there is provided a single domain antibody comprising a heavy chain variable region comprising CDRs 1-3.
Wherein i) CDR1 comprises SEQ ID NO:34, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:35, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:36, and a nucleotide sequence shown in seq id no. Or ii) CDR1 comprises SEQ ID NO:37, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:38, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:39, and a polypeptide having the amino acid sequence shown in seq id no.
In one embodiment of the invention, the single domain antibody comprises SEQ ID NO:28 or 29.
In a sixth aspect of the present invention, there is provided a method for producing the above antibody or antigen-binding fragment or the above single domain antibody, the method comprising: preparing a phage display library, and screening the phage display library to obtain antibodies or antigen binding fragments or single domain antibodies.
In a seventh aspect of the invention, there is provided an antigen receptor comprising a transmembrane region, an intracellular region and one or more identical or different extracellular binding domains.
The antigen receptor is a TCR or CAR.
In one embodiment of the invention, the antigen receptor is a CAR.
The extracellular binding domain is an extracellular antigen binding domain, an extracellular antibody binding domain, a receptor and a ligand, wherein the receptor is preferably a natural T cell receptor.
In one embodiment of the invention, the extracellular binding domain is an extracellular antigen binding domain.
The extracellular antigen-binding domain is derived from an antibody.
Preferably, the transmembrane region is linked directly or via a linker to one or more extracellular antigen binding domains.
Preferably, the antigen is selected from cancer-related antigens, for example from the group consisting of: GPC3, CD16, CD64, CD78, CD96, CLL1, CD116, CD117, CD71, CD45, CD71, CD123, CD138, erbB2 (HER 2/neu), carcinoembryonic antigen (CEA), epithelial cell adhesion molecule (EpCAM), epidermal Growth Factor Receptor (EGFR), EGFR variant III (EGFRvIII), CD19, CD20, CD30, CD40, bissialoglycganglioside GD2, ductal mucin, gp36, TAG-72, glycosphingolipids, glioma-associated antigen, beta-human chorionic gonadotrophin, alpha Fetoprotein (AFP), lectin-reactive AFP, thyroglobulin, RAGE-1, MN-CA IX, human telomerase reverse transcriptase, RU1, RU2 (AS), enterocarboxylesterase, mut hsp70-2, M-CSF prostase (prostase), prostase Specific Antigen (PSA), PAP, NY-ESO-1, LAGA-1a, p53, prostein, PSMA, survival and telomerase, prostate cancer tumor antigen-1 (PCTA-1), MAGE, ELF2M, neutrophil elastase, ephrin B2, CD22, insulin growth factor (IGF 1) -I, IGF-II, IGFI receptor, mesothelin, major Histocompatibility Complex (MHC) molecules presenting tumor specific peptide epitopes, 5T4, ROR1, nkp, NKG2D, tumor matrix antigen, additional domain a (EDA) and additional domain B (EDB) of fibronectin, A1 domain of tenascin-C (TnC A1), fibroblast-related protein (fap), CD3, CD4, CD8, CD24, fibronectin, CD25, CD33, CD34, CD133, CD138, foxp3, B7-1 (CD 80), B7-2 (CD 86), GM-CSF, cytokine receptor, endothelial factor, major Histocompatibility Complex (MHC) molecule 、BCMA(CD269、TNFRSF17)、TNFRSF17(UNIPROTQ02223)、SLAMF7(UNIPROT Q9NQ25)、GPRC5D(UNIPROT Q9NZD1)、FKBP11(UNIPROT Q9NYL4)、KAMP3、ITGA8(UNIPROT P53708), or FCRL5 (UNPROT Q68SN 8);
Preferably, the antigen is mesothelin.
Preferably, the extracellular antigen-binding domain comprises CDR1-3, wherein i) CDR1 comprises SEQ ID NO:34, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:35, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:36, and a nucleotide sequence shown in seq id no. Or ii) CDR1 comprises SEQ ID NO:37, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:38, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:39, and a polypeptide having the amino acid sequence shown in seq id no.
In one embodiment of the invention, the extracellular antigen-binding domain comprises an antibody or antigen-binding fragment as described above, a single chain antibody as described above, or a single domain antibody as described above.
Preferably, the transmembrane region is derived from human CD8.
Preferably, the intracellular domain is derived from 4-1BB, CD28 or CD3 zeta.
In an eighth aspect of the invention, there is provided a nucleic acid encoding the STAR, the STAR complex, the antibody or antigen binding fragment, the single chain antibody, the single domain antibody, the antigen receptor.
In a ninth aspect of the invention, there is provided a vector comprising a nucleic acid as described above.
The vector can be expressed in vivo or in vitro or ex vivo, preferably an expression vector. Preferably, the expression vector is a prokaryotic expression vector, a viral expression vector, a plasmid, a cosmid, a phage, a virus, or the like.
Preferably, the prokaryotic expression vector is escherichia coli series. Such as pET-26b or pET28a+.
Preferably, it may be Rous Sarcoma Virus (RSV), lentivirus, human Immunodeficiency Virus (HIV), murine Leukemia Virus (MLV), equine Infectious Anemia Virus (EIAV), mouse breast cancer virus (MMTV), fujinami sarcoma virus (FuSV), FBR murine osteosarcoma virus (FBR MSV), moloney murine leukemia virus (Mo-MLV), moloney murine sarcoma virus (Mo-MSV), abelson murine leukemia virus (A-MLV), avian myeloproliferative virus 29 (MC 29) or avian myeloblastosis virus (AEV), etc. Still more preferably, the expression vector is a lentiviral expression vector. Such as pHAGE-IRES-RFP.
In a tenth aspect of the invention, there is provided a host cell comprising a nucleic acid as described above or a vector as described above.
Preferably, the host cell may be eukaryotic or prokaryotic. More preferably, the host cell is a yeast cell, 293 cell, CHO cell, escherichia coli, or the like.
In an eleventh aspect of the invention, an immune cell is provided, said immune cell expressing the STAR, the STAR complex, the antibody or antigen binding fragment, the single domain antibody, the antigen receptor.
Preferably, the immune cell comprises one or more of the above-described nucleic acids.
Preferably, the immune cells are selected from T cells, treg cells, macrophages, NK cells, NKT cells, peripheral blood mononuclear cells, TIL cells or Dendritic Cells (DCs).
Preferably, the immune cells are derived from T cells of the subject.
In one embodiment of the invention, the immune cells are selected from T cells, NK cells, CTLs, human embryonic stem cells, lymphoid progenitor cells and/or T cell precursor cells.
In a twelfth aspect of the invention, there is provided a CAR-T cell comprising an antibody or antigen fragment as described above, a single chain antibody as described above or a single domain antibody as described above.
According to a thirteenth aspect of the present invention, there is provided a method for producing an immune cell, which comprises transfecting the above-described nucleic acid sequence into an immune cell to express the nucleic acid sequence.
In a fourteenth aspect of the present invention, there is provided a method for preparing a recombinant T cell, comprising the steps of:
1) Cloning from positive T cells to obtain the nucleic acid described above;
2) Isolating and culturing primary T cells;
3) Delivering the nucleic acid obtained in step 1) into the primary T cells described in step 2) to obtain recombinant T cells expressing STAR as described above.
In a fifteenth aspect of the present invention, there is provided a method for preparing a STAR or a STAR complex, comprising the steps of:
(1) Cloning from positive T cells to obtain the nucleic acid described above;
(2) Connecting the nucleic acid obtained in the step (1) to a vector skeleton to obtain an expression vector;
(3) Transforming the expression vector obtained in the step (2) into a host cell, and then inducing the expression thereof;
(4) STAR is obtained.
In a sixteenth aspect of the invention, there is provided a method of producing an antibody or antigen-binding fragment, single chain antibody or single domain antibody, said method comprising protein and/or DNA immunization.
In a seventeenth aspect of the present invention, there is provided a method for producing an antibody or antigen-binding fragment, single chain antibody or single domain antibody, said method comprising:
A) Obtaining a coded nucleic acid sequence;
b) Transforming the nucleic acid sequence obtained in the step A) into a host cell, and then inducing the expression and purification of the host cell.
In an eighteenth aspect of the invention, there is provided the use of a STAR as defined above, a STAR complex as defined above, an antibody or antigen binding fragment as defined above, a single domain antibody as defined above, an antigen receptor as defined above, a nucleic acid as defined above, or an immune cell as defined above for the preparation of a product for the diagnosis or treatment of a tumor.
Preferably, the neoplasm includes, but is not limited to, lymphoma, non-small cell lung cancer, leukemia, ovarian cancer, nasopharyngeal cancer, breast cancer, endometrial cancer, colon cancer, rectal cancer, gastric cancer, bladder cancer, lung cancer, bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, liver and bile duct cancer, esophageal cancer, renal cancer, thyroid cancer, head and neck cancer, testicular cancer, glioblastoma, astrocytoma, melanoma, myelodysplastic syndrome, and sarcoma. Wherein the leukemia is selected from acute lymphoblastic (lymphoblastic) leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, multiple myeloma, plasma cell leukemia, and chronic myelogenous leukemia; the lymphoma is selected from hodgkin's lymphoma and non-hodgkin's lymphoma, including B-cell lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone B-cell lymphoma, T-cell lymphoma, and waldenstrom's macroglobulinemia; the sarcoma is selected from osteosarcoma, ewing sarcoma, leiomyosarcoma, synovial sarcoma, soft tissue sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma, and chondrosarcoma.
In a nineteenth aspect of the invention, there is provided an immunoconjugate or antibody drug conjugate comprising an antibody or antigen-binding fragment of the invention as described above, a single chain antibody as described above, or a single domain antibody as described above conjugated to a therapeutic or diagnostic agent.
In a twentieth aspect of the present invention, a pharmaceutical composition comprising the STAR, the STAR complex, the antibody or antigen binding fragment, the single domain antibody, the antigen receptor, the nucleic acid, and the immune cell is provided.
Preferably, the medicament further comprises pharmaceutically acceptable auxiliary materials. Further preferably, the pharmaceutically acceptable excipients include, but are not limited to, diluents, binders, wetting agents, surfactants, lubricants or disintegrants, and the like.
In a twenty-first aspect of the present invention, a kit is provided, said kit comprising the STAR, the STAR complex, the antibody or antigen binding fragment, the single domain antibody, the antigen receptor, the nucleic acid, and the immune cell.
In a twenty-second aspect of the invention, there is provided a method of treating a tumor, the method comprising administering to a subject an effective amount of a STAR, STAR complex, CAR, antibody or antigen binding fragment thereof, single chain antibody, single domain antibody, immune cell, CAR-T cell or pharmaceutical composition of the invention.
"Linker" as used herein includes, but is not limited to, rigid linkers, flexible linkers, cleavable linkers, or nonsensical amino acids. Preferably, the amino acid sequence of the rigid linker is selected from the group consisting of SEQ ID NO:49-59, and one or more than two thereof. Preferably, the flexible linker is selected from glycine and/or serine rich peptide fragments; preferably, the flexible linker is selected from the group consisting of SEQ ID NO:60-112, wherein said cleavable linker is selected from the group consisting of SEQ ID NO: 113-117.
The "antibodies" described herein may be of any class (e.g., igA, igD, igE, igG and IgM) or subclass (e.g., igG1, igG2, igG3, igG4, igA1, or IgA 2).
The "antigen binding fragments" of the present invention include, but are not limited to: a Fab fragment having VL, CL, VH and CH1 domains; a Fab' fragment, which is a Fab fragment having one or more cysteine residues at the C-terminus of the CH1 domain; fd fragment with VH and CH1 domains; fd' fragments having VH and CH1 domains and one or more cysteine residues at the C-terminus of the CH1 domain; fv fragments having the VL and VH domains of a single arm of an antibody; a dAb fragment consisting of a VH domain or a VL domain; an isolated CDR region; a F (ab ') 2 fragment, which is a bivalent fragment comprising two Fab' fragments linked by a disulfide bridge at the hinge region; single chain antibody molecules (e.g., single chain Fv; scFv); a "diabody" having two antigen binding sites comprising a heavy chain variable domain (VH) linked to a light chain variable domain (VL) in the same polypeptide chain; a "linear antibody" comprising a pair of tandem Fd segments (VH-CH 1-VH-CH 1) that form a pair of antigen-binding regions with complementary light chain polypeptides; and modified versions of any of the foregoing that retain antigen binding activity.
"CDR" as used herein refers to complementarity determining regions within the variable sequences of an antibody. For each variable region, there are three CDRs in each of the heavy and light chains, which are referred to as CDR1, CDR2, and CDR3. The exact boundaries of these CDRs are defined differently for different systems. The system described by Kabat et al (Kabat et al,Sequences of Proteins of Immunological Interest(National Institutes of Health,Bethesda,Md.(1987) and (1991)) provides not only a well-defined residue numbering system for the variable regions of antibodies, but also residue boundaries defining the three CDRs. These CDRs may be referred to as Kabat CDRs. Each complementarity determining region may comprise amino acid residues from a "complementarity determining region" as defined by Kabat. Chothia et al (Chothia & Lesk, J.mol. Biol,196:901-917 (1987) and Chothia et al, nature 342:877-883 (-1989)) found that some of the sub-portions within the Kabat CDRs employed nearly identical peptide backbone conformations, albeit with great diversity at the amino acid sequence level. These subfractions are referred to as L1, L2 and L3 or H1, H2 and H3, respectively, wherein "L" and "H" represent the light and heavy chain regions, respectively. These regions may be referred to as Chothia CDRs, which have boundaries that overlap with Kabat CDRs. Still other CDR boundary definitions may not strictly follow one of the above systems, but will still overlap with Kabat CDRs, and the methods used herein may utilize CDRs defined according to any of these systems, although preferred embodiments use Kabat or Chothia defined CDRs. The "antibody variable region" refers to a portion of the light and heavy chains of an antibody molecule that includes the amino acid sequences of complementarity determining regions (CDRs, i.e., CDR1, CDR2, and CDR 3) and Framework Regions (FR). VH refers to the variable domain of the heavy chain. VL refers to the variable domain of the light chain.
The term "diagnosis" as used herein refers to ascertaining whether a patient has a disease or condition in the past, at the time of diagnosis, or in the future, or ascertaining the progression or likely progression of the disease in the future, or assessing the patient's response to treatment.
"Treatment" as used herein means slowing, interrupting, arresting, controlling, stopping, alleviating, or reversing the progression or severity of a sign, symptom, disorder, condition, or disease, but does not necessarily refer to the complete elimination of all disease-related signs, symptoms, conditions, or disorders, and refers to therapeutic intervention to ameliorate the signs, symptoms, etc. of a disease or pathological condition after the disease has begun to develop.
An "effective amount" as used herein refers to an amount or dose of STAR, STAR complex, CAR-T, STAR-T, immune cells, pharmaceutical compositions, etc. as described herein that provides the desired treatment or prevention after administration to a patient or organ in single or multiple doses.
The "product" of the present invention may be a kit, a chip, an antibody conjugate, a multifunctional antibody, a pharmaceutical composition, or the like.
The "individual" or "subject" of the present invention may be a human or non-human animal, and the non-human animal may be a non-human mammal such as a mouse, a cow, a sheep, a rabbit, a pig, a monkey, etc.
All combinations of items to which the term "and/or" is attached "in this description shall be considered as being individually listed in this document. For example, "a and/or B" includes "a", "a and B", and "B". Also for example, "A, B and/or C" include "a", "B", "C", "a and B", "a and C", "B and C" and "a and B and C".
The term "comprising" or "comprising" as used herein is an open reading frame, and when used to describe a sequence of a protein or nucleic acid, the protein or nucleic acid may consist of the sequence, or may have additional amino acids or nucleotides at one or both ends of the protein or nucleic acid, but still have the activity described herein. Furthermore, it will be clear to those skilled in the art that the methionine encoded by the start codon at the N-terminus of a polypeptide may be retained in some practical situations (e.g., when expressed in a particular expression system) without substantially affecting the function of the polypeptide. Thus, in describing a particular polypeptide amino acid sequence in the present specification and claims, although it may not comprise a methionine encoded at the N-terminus by the initiation codon, a sequence comprising such methionine is also contemplated at this time, and accordingly, the encoding nucleotide sequence may also comprise the initiation codon; and vice versa.
Embodiments of the present invention are described in detail below with reference to the attached drawing figures, wherein:
Fig. 1: antibody expression and specificity identification of NM5/NM 24-VHH. Wherein, FIG. 1A shows the antibody expression of NM5-VHH, and M is Marker. FIG. 1B shows the antibody expression of NM24-VHH, M is Marker. FIG. 1C shows the results of specific staining after incubation of antibodies with AsPC-1 cells. FIG. 1D shows the results of specific staining of antibodies after co-incubation with 293T-MSLN cells, PC-Ab for publicly known MSLN antibodies, positive control, NC for negative control.
Fig. 2: schematic representation of optimization of STAR by constant region cysteine modification, transmembrane and intracellular region modification.
Fig. 3: schematic representation of optimization of STAR by addition of co-stimulatory molecule receptor intracellular domains to the alpha and/or beta chain.
Fig. 4: schematic representation of optimization of STAR by deletion of the intracellular regions of the alpha and/or beta chains followed by addition of co-stimulatory molecule receptor intracellular domains either directly or through linkers.
Fig. 5: schematic representation of STAR optimization by addition of co-stimulatory molecule receptor intracellular domains to the CD3 subunit.
Fig. 6: schematic representation of optimization of STAR by addition of cytokine receptor intracellular signaling regions to the alpha and/or beta chain.
Fig. 7: affinity detection of MSLN single domain antibodies NM5 (a) and NM24 (B).
Fig. 8: the anti-MSLN-co-STAR structure is schematically shown.
Fig. 9: experimental results of infection efficiency (rfp+ subgroup ratio) of NM5, NM13, NM24 STAR and Negative Control (NC).
Fig. 10: target cells expressing MSLN were selected, a being pancreatic cancer tumor cell line, B being antigen overexpressing cell line.
Fig. 11: comparison of killing function in vitro for different VHHs. AsPC-1 belongs to a human metastatic pancreatic adenocarcinoma cell line and is a MSLN positive target cell. 293T was transfected to express human hMSLN and murine mMSLN, respectively. FIG. 11A shows in vitro killing of various nanobodies on AsPC-1 and 293T cells, FIG. 11B shows that STAR T cells of four nanobody sequences NM5, 11, 13, 24 have a strong killing effect on Aspc-1, and FIG. 11C shows that NM5 and NM24 STAR T cells are capable of recognizing and killing 293T-hMSLN but not 293T-mMSLN target cells.
Fig. 12: cytokine release levels of anti-MSLN STAR comprehensive mutants based on NM5/NM 24-VHH. Of these, FIG. 12A shows IFN- γ release levels, FIG. 12B shows IFN- γ release levels of NM5 STAR T cells, NM11 STAR T cells, NM13 STAR T cells, NM24STAR T cells after 24 and 48 hours, FIG. 12C shows IL-2 release levels, and FIG. 12D shows IL-2 release levels of NM5 STAR T cells, NM11 STAR T cells, NM13 STAR T cells, NM24STAR T cells after 24 and 48 hours.
Fig. 13: an anti-MSLN-STAR-T cell tumor treatment result graph, wherein FIG. 13A is an in vivo anti-tumor effect of Mock T cells, NM5 STAR T cells, NM13 STAR T cells and NM24 STAR T cells in a mouse tumor model; FIG. 13B is a graph showing tumor fluorescence statistics in mice corresponding to FIG. 13A; FIG. 13C is the change in body weight of mice after back transfusion of Mock T cells, NM5 STAR T cells, NM13 STAR T cells, NM24 STAR T cells; FIG. 13D shows proliferation of Mock T cells, NM5 STAR T cells, NM13 STAR T cells, NM24 STAR T cells in mice; FIG. 13E shows the percentage of CD4 + T cells in a mouse tumor model for Mock T cells, NM5 STAR T cells, NM13 STAR T cells, NM24 STAR T cells; FIG. 13F shows the percentage of CD8 + T cells in a mouse tumor model for Mock T cells, NM5 STAR T cells, NM13 STAR T cells, NM24 STAR T cells.
Fig. 14: an anti-MSLN-STAR-T cell tumor therapeutic outcome graph, wherein fig. 14A is an in vivo anti-tumor effect of the constructed anti-MSLN-STAR-T cells in a mouse tumor model when (G4S) nlinker is n=0 and n=5, respectively; FIG. 14B is a graph of in vivo tumor fluorescence statistics of mice corresponding to graph A; FIG. 14C shows proliferation of integrated variants of anti-MSLNSTAR constructed from α -del-OX40 and α -del- (G4S) 5-OX40 in mice; FIG. 14D is the percentage of CD62L +CD8 + T cells in a mouse tumor model for the anti-MSLN STAR integrated variants constructed with alpha-del-OX 40 and alpha-del- (G4S) 5-OX 40.
Fig. 15: pharmacokinetic analysis experiments of NM24 STAR-T cells in tissues returned for 0-21 days, wherein FIG. 15A is the copy number of NM24 STAR T cells; FIG. 15B is the percentage of NM24 STAR T cells.
Fig. 16: in vitro killing and IFN-gamma secretion of NM24 STAR-T cells and similar cell products TCR 2 TC-210 (SD 1-eTruC T cells) are compared, wherein FIG. 16A shows the in vitro killing effect, and FIG. 16B shows the release level of IFN-gamma secretion, wherein E/T ratio is the effective target ratio.
Fig. 17: the in vivo efficacy and safety of NM24 STAR-T cells were compared with those of similar cell products TCR 2 TC-210 (SD 1-eTruC T cells), wherein FIG. 17A shows the in vivo anti-tumor effects of Mock T cells, NM24 STAR T cells, SD1-eTruC T cells in a mouse tumor model; FIG. 17B is a graph showing tumor fluorescence statistics in mice corresponding to FIG. 17A; FIG. 17C is the change in body weight of mice after back transfusion of Mock T cells, NM24 STAR T cells, SD1-eTruC T cells; FIG. 17D shows the survival rate of mice after back transfusion of Mock T cells, NM24 STAR T cells, SD1-eTruC T cells.
The technical solutions of the embodiments of the present invention will be clearly and completely described below with reference to the drawings in the embodiments of the present invention, and it is apparent that the described embodiments are only some embodiments of the present invention, not all embodiments. All other embodiments, which can be made by those skilled in the art based on the embodiments of the invention without making any inventive effort, are intended to be within the scope of the invention.
Example 1: single domain antibody sequence screening targeting MSLN
1. Preparation of human MSLN proteins
1) Construction of expression vectors
The cDNA of human MSLN high expression tumor cell is used as template, the DNA sequence of human MSLN protein extracellular region is shown as SEQ ID No.120, and the amino acid sequence is shown as SEQ ID No. 121. The extracellular domain of MSLN was obtained by PCR amplification. The amplified MSLN extracellular region is connected to pVRC expression vector by enzyme cutting-connecting method, and is transformed into DH5 alpha competent cell, single colony is extracted or plasmid is extracted, and sequencing is carried out.
2) Protein expression
Transfection was performed when 293F cell densities were as high as about 2X 10 6 cells/mL, 200. Mu.g of DNA was required to transfect 200mL cells, and the mass ratio of plasmid DNA to polyethyleneimine PEI (concentration 1 mg/mL) was about 1:3, 200 μg:600 μg, plasmid DNA and PEI were each added to a final volume of 5mL of non-resistant medium. The PEI and medium mixture was added to the plasmid DNA and medium mixture and incubated for about 15-30min, the volume of the incubation mixture being about 1/10 of the volume of the cells to be transfected. And adding the incubated mixed solution into 293F cells, culturing for 72-96h, and collecting cells or culture medium supernatant for protein extraction.
3) Protein purification
The collected culture supernatant was centrifuged to remove the precipitate, and the supernatant was filtered through a 0.22 μm filter membrane and concentrated and replaced with a lysate using a concentrate bag. Washing 10 column volumes of nickel columns with lysate washing liquid respectively to remove nonspecifically bound impurity proteins, and eluting target proteins from the affinity chromatography column with eluent. After concentration of the displacement solution, the concentration was determined, the samples were frozen and left for SDS-PAGE.
2. Human MSLN protein immune alpaca
Human MSLN protein extracellular region (100 μg) expressed and purified using the eukaryotic system at step 1 above was used to immunize healthy alpaca, and adjuvants include complete Freund's adjuvant (CFA, sigma) and incomplete Freund's adjuvant (IFA, sigma). The extracellular domain of the expressed and purified human MSLN protein was diluted with PBS and then mixed with the corresponding adjuvant 1:1. The antigen and adjuvant were thoroughly mixed to form a stable emulsion, the antigen mixture was withdrawn with a syringe and injected subcutaneously under the skin of the alpaca neck in multiple spots of 100-200 μl each. The specific immunization animal procedure is as follows:
1) Day 1 (immunization 1): immunization of alpaca using Complete Freund's Adjuvant (CFA) mixed MSLN antigen (100 μg) subcutaneously;
2) Day 14 (immunization 2): immunization of alpaca using Incomplete Freund's Adjuvant (IFA) mixed MSLN antigen (100 μg) subcutaneously;
3) Day 28 (immunization 3): the alpaca was immunized again by subcutaneous injection with Incomplete Freund's Adjuvant (IFA) mixed MSLN antigen (100 μg);
4) Day 42 (serum 1): taking alpaca blood samples from alpaca veins, extracting serum, and detecting antibody titer, wherein the P/N value of the serum diluted by 20 ten thousand times is more than 2;
5) Day 42 (immunization 4): subcutaneously injecting MSLN antigen (100 mug) mixed with Incomplete Freund's Adjuvant (IFA) again to immunize alpaca and enhance the immune effect;
6) Day 53 (serum 2): taking alpaca blood samples from alpaca veins, extracting serum, and detecting antibody titer, wherein the P/N value of the serum diluted by 20 ten thousand times is more than 2;
7) Day 54, 57, 60: 30-40mL alpaca blood samples were taken from the alpaca hindleg vein and PBMC isolation was performed.
PBMC isolation
1) PBMCs were isolated in a biosafety cabinet, and the blood in the anticoagulation tube was combined into one tube into 50mL centrifuge tube (30 mL), PBS was added to a total volume of 50mL, and gently mixed.
2) A new 50mL centrifuge tube was added with Ficoll separation solution, 15mL/tube. A blood sample, 25mL/tube, was placed on the Ficol level. The operation process is stable, and the layering of Ficoll and blood is formed to prevent mixing.
3) The lifting speed of the centrifuge is adjusted to 0, RT, centrifugal force is 800g, and the centrifuge is centrifuged for 30min. After centrifugation, the sample was removed from the centrifuge and the samples were stratified as follows: upper aqueous phase-buffy coat-Ficoll layer-erythrocyte layer, wherein PBMC are in the buffy coat, and the buffy coat is aspirated and transferred to a new 50mL centrifuge tube.
4) PBS was added to the sample tube to 50mL, mixed well, centrifuged at 2000rpm, at RT for 5min, the supernatant was discarded, and 5mL PBS was added to resuspend the cell pellet.
5) And (4) repeating the step (4) and simultaneously cooling to 4 ℃.
6) Cell pellet was resuspended in 5mL PBS, added PBS to 40mL, and cell counted.
7) Centrifuging at 1500rpm at 4deg.C for 5min, discarding supernatant, adding 1mL PBS, resuspending cells, and blowing well; adding 20mL Trizol, mixing, standing at room temperature for 5min, lysing cells, and sub-packaging into 1.5mL EP tube of RNase-free, and freezing at-80deg.C for extracting RNA.
4. RNA extraction and reverse transcription
1) The sample was taken out from-80 ℃, thawed at room temperature, 200. Mu.L of chloroform was added, and left standing at room temperature for 3min. The sample was centrifuged at 12000g for 15min at 4℃and separated into an upper aqueous phase, an intermediate layer and an organic layer, the upper layer was transferred to a new RNase-free tube, 1. Mu.L glycogen was added, and isopropanol was added in a proportion of 500. Mu.L isopropanol, and the mixture was allowed to stand at 4℃overnight.
2) The sample was centrifuged at 12000g at 4℃for 20min, the supernatant was aspirated, 1mL of pre-chilled 75% alcohol was added to wash the pellet, the supernatant was again centrifuged to aspirate alcohol, and the pellet was dried. RNA pellet was dissolved in 15. Mu.L of RNase-free water per tube, and reverse transcription was performed.
3) CDNA synthesis (20. Mu.L system) was performed using Promega reverse transcription kit.
The first step: taking a certain template RNA, and adding Oligo (dT), wherein the table 1 is shown;
TABLE 1
Component (A) | Volume (mu L) |
RNA(<5μg/reaction) | 11 |
Oligo(dT)15Primer | 1 |
Total volume of | 12 |
And a second step of: the mixture of template RNA and Oligo (dT) was pre-denatured at 65℃for 5min, and placed back on ice after completion.
And a third step of: in the case of pre-denaturation, RT-Mix was prepared in advance, 8. Mu.L per tube, and the composition and volume are shown in Table 2.
TABLE 2
Component (A) | Volume (mu L) |
5×Reaction Buffer | 4μL |
RiboLockRnaseInhibitor(20U/μL) | 1μL |
10mM dNTP Mix | 2μL |
RevertAid M-MuLVRT(200U/μL) | 1μL |
Total volume of | 8μL |
Fourth step: reverse transcription program is set, and extension and reverse transcriptase are inactivated. After the procedure was completed, cDNA was obtained.
5. Phage library construction
1) PCR acquisition of VHH sequences
The VHH sequence was obtained by two rounds of PCR and homology arms of the vector were added to both ends of the sequence.
2) First round PCR
Step 1: the preparation reaction system is shown in Table 3.
TABLE 3 Table 3
Step 2: the PCR conditions are shown in Table 4.
TABLE 4 Table 4
The PCR product was subjected to gel electrophoresis, and the target band at 0.7kb was excised and the product was recovered.
3) Second round PCR
Step 1: the preparation reaction system is shown in Table 5.
TABLE 5
Step 2: the PCR conditions are shown in Table 6.
TABLE 6
And (3) carrying out gel electrophoresis on the PCR product, cutting a 400bp target band, and recovering the product.
4) Vector PCR
The vector region of phagemid was obtained by PCR for expression of VHH sequences.
Step 1: the preparation reaction system is shown in Table 7.
TABLE 7
Step 2: the PCR conditions are shown in Table 8.
TABLE 8
The PCR product is subjected to gel electrophoresis, a 4000bp target band is cut, and the product is recovered.
5) Ligation, ligation product purification and concentration
The VHH fragment was ligated to phagemid vector and the ligation product was subsequently concentrated.
Step 1: the preparation reaction system is shown in Table 9.
TABLE 9
Step 2: the mixture was incubated at 50℃for 2h and cooled on ice.
Step 3: purifying the connection product, removing salt ions, proteins and other components in the connection system, and concentrating the volume to 1/10 of the original volume.
6. Electric transfer warehouse
1) One tube of E.coli was competent and thawed on ice.
1) Mu.L of the ligation product or positive control was added to the competence and gently blown. Standing on ice for 1-2min, transferring to a precooled electric rotating cup, and performing electric rotation.
2) After electrotransformation, 1mL of 2YT-G at 37 ℃ is immediately added, a gun head is used for flushing an electrotransformation cup, bacterial liquid after electrotransformation is transferred into a 15mL centrifuge tube or a 2mL EP tube, and water bath at 37 ℃ is recovered until all samples are electrotransformed. (2 YT-G: 2 XYT medium containing 2% glucose) was transferred to a shaking table at 37℃and recovered at 220rpm for 1 hour.
3) Mu.L of the bacterial liquid was aspirated from the bacterial liquid, diluted 10-5 times, plated with 2YT-A (2 YT plate containing 100. Mu.g/mL ampicillin) and incubated overnight at 37℃in an incubator for colony counting.
4) The rest bacterial liquid is inoculated into a 2YT-AG culture medium, shaken to a logarithmic phase, added with auxiliary phage for infection, and shaken at 30 ℃ and 220rpm for 12-16 hours. ( 2YT-AG: 2 XYT medium containing 2% Glucose and 100. Mu.g/mL ampicillin )
5) Phage were harvested and concentrated and titers were determined.
7. Phage library antibody screening
Three rounds of antibody screening are carried out on the phage library obtained in the step 7, and each screening comprises one positive selection and one negative selection. The phage were incubated with the antigen peptide and the unbound phage was discarded, leaving phage bound to the antigen peptide, and then incubated with BSA for negative selection, leaving phage unbound to BSA.
1) Coat plate. The antigen was diluted to a concentration of 2 ng/. Mu.L with PBS and added to a 96-well plate at 100. Mu.L/well; 2% BSA was prepared with PBS and added to the corresponding wells, 100. Mu.L/well, sealed with preservative film and incubated overnight at 4 ℃.
2) The coating solution was discarded, and 200. Mu.L of a washing solution (washing solution: 1% Tween 20/PBS, pH 7.4) and washed 3 times.
3) And (5) sealing. All wells were added with 2% BSA blocking solution, 100. Mu.L/well, sealed with preservative film and incubated at 37℃for 1h.
4) The supernatant was discarded, and 200. Mu.L of the washing solution was added thereto, followed by washing 3 times.
5) Phage were added to the wells of the positive selection, diluted to 100. Mu.L in volume at 1X 10 12 pages/well, sealed with preservative film and incubated for 1h at 37 ℃.
6) The supernatant was discarded, 200. Mu.L of wash solution was added, and the mixture was washed 10 times.
7) Elution-neutralization. 200. Mu.L of eluent was added to the wells of the cation selection and neutralized to pH 7-7.4.
8) Selecting yin. And adding the eluent into the anion-selective holes, sealing by using a preservative film, incubating for 1h at 37 ℃, sucking and retaining the supernatant, and detecting the titer.
9) A small amount of phage after Panning rounds of dilution is coated on a2 XYT-A plate at 37 ℃ overnight; counting the colonies the next day, and calculating the titer; and monoclonal sequencing is selected, and sequence diversity and enrichment conditions are analyzed.
10 All remaining phages were used for TG1 infection.
11 M13KO7 infection. Diluting phage, adding M13KO7 into the bacterial liquid, and carrying out water bath at 37 ℃ for 30min; 2 XYT-AK medium was changed, and shaking was performed at 30℃and 220rpm for 14-16 hours.
12 Concentrating phage and detecting phage titer, then the next round of screening can be performed.
The amount of coated antigen of two-round screening and three-round screening is reduced, the washing times are increased after the phage and the positive hole are incubated, and other steps are the same as the above steps.
8. Binding detection and sequence acquisition
The phage obtained by three rounds of screening and M13KO7 auxiliary phage are jointly infected with TG1, a 2YT-AK plate is coated, monoclonal is selected for phage expansion, phage is collected for combination detection, and available phage/antibodies are determined.
1) Coat plate. Diluting the antigen with the coating solution to 1 ng/. Mu.L, 100. Mu.L/well; control negative wells were added with 2% bsa; sealing with fresh-keeping film, and standing at 4deg.C overnight.
2) The coating solution in the plate was discarded, and 200-250. Mu.L of wash solution was added to wash 3 times.
3) 200. Mu.L of 2% BSA was added to all wells and blocked at room temperature for 1h.
4) The blocking solution in the plate was discarded, 200. Mu.L of wash solution was added, and the solution was washed 1 time.
5) 100. Mu.L of phage was added to each of the positive and negative wells and incubated at 37℃for 1h.
6) Phage in the plates were discarded, 200. Mu.L of wash was added and washed 3 times.
7) The anti-M13-HRP antibody was diluted, 50ng/well and incubated for 1h at room temperature.
8) The antibody in the plate was discarded, 200. Mu.L of wash solution was added, and the solution was washed 5 times.
9) 100 Mu L of TMB color development liquid is added into each hole, and the reaction is carried out at room temperature until the OD value is between 2 and 3.
10 Stop solution was added to the color development system at 50. Mu.L per well.
11A spectrophotometric measurement of the absorbance at 450 nm. And (5) sequencing the monoclonal bacterial liquid corresponding to the positive hole, and determining the VHH sequence.
12 The antibodies obtained were designated NM1-24, respectively.
9. Cell level functional screening
1) Assembling the positive antibody VHH sequence obtained by screening with a STAR molecule constant region, and inserting a lentiviral vector by using a homologous recombination method to construct a complete STAR plasmid.
2) Packaging virus
Lentix-293T cells were inoculated into 10cm dishes at 5X 10 5/mL, incubated in a 5% CO 2 incubator at 37℃and transfected when cell density reached about 80% (observed under a microscope). The four plasmids were homogeneously mixed with 500. Mu.L of serum-free DMEM in a PMD2.G: PRSV-Rev: PMDlg: TRANSFER PLAMID =1:1:2:4 ratio. mu.L of PEI-Max was mixed with 500. Mu.L of serum-free DMEM and allowed to stand at room temperature for 5min (PEI-Max to plasmid volume/mass ratio 3:1). The PEI-max mixture was slowly added to the plasmid mixture, gently swirled, mixed well and allowed to stand at room temperature for 15min. And slowly adding the final mixed solution into a culture medium, fully and uniformly mixing, putting the mixture back into an incubator for continuous culture for 12-16 h, changing the culture medium into a 6% FBS DMEM culture medium for continuous culture, and collecting the virus liquid for 48h and 72 h.
3) Virus titer measurement
The TCR knocked-out Jurkat-C4 cells were seeded at 1.5X10 5/mL in flat bottom 96-well plates, and 100. Mu.L of 1640 medium containing 10% FBS, 0.2. Mu.L of 1000 Xpolybrene was added to each well. The virus was diluted 10-fold with 1640 complete medium. Adding diluted cells into a virus hole, mixing, centrifuging for 90min at 32 ℃ and 1500rpm, culturing in a 5% CO 2 incubator at 37 ℃ for 72 hours, measuring infection efficiency by a flow cytometer, and selecting a hole with the infection rate of 2-30% when the titer is calculated, wherein the calculation formula is as follows: titer (TU/mL) =1.5X10≡4×positive rate ≡viral volume (. Mu.L) ×1000. The above viruses were infected with T cells and STAR was expressed.
4) Isolation, activation and infection of human primary T cells
After primary T cells were obtained by Ficoil isolation, they were cultured in X-VIVO medium containing 10% FBS and 100IU/mL IL-2 at an initial culture density of 1X 10 6/mL and activated by addition to CD3, CD28 and fibractin pre-coated well plates. After 24h activation, adding the virus liquid, centrifuging at 1500rpm for 90min, and culturing in a CO 2 incubator. X-VIVO medium containing 10% FBS and 100IU/mL IL-2 was supplemented 24h after infection and plated out, and subcultured at 1-2 days later.
5) Determination of killing efficiency and cytokine Release by in vitro Co-culture of T cells and target cells
Target cells AsPC-1 or 293T cells overexpressing MSLN are plated in 24-well plates overnight at a density of 1E 5/well and cultured, and corresponding numbers of STAR-T cells are added to the target cells according to the ratio of STAR-positive T cells to target cells of 3:1, 1:1 and 1:3, and the killing efficiency of the STAR-T cells on the target cells and the concentration of cytokines released from the supernatant after 24 hours or 48 hours of culture are detected.
10. Antibody expression and specificity identification
The VHH regions of NM5 and NM24 are fused with the Fc region of human IgG1 to construct plasmids, the plasmids are transferred into 293F cells, and the supernatants are collected and purified to obtain the corresponding antibodies. And the antibody size was identified by SDS-PAGE. And (3) staining and flow-type determination are carried out on 293T-MSLN cells by using the purified antibody, so as to determine the specificity of the antibody. (see FIGS. 1A-1D)
11. Antibody affinity assay
The CM5 chip is used for fixing MSLN protein as a stationary phase, the recombinant expressed antibody NM5-hFc or NM24-hFc is used as a mobile phase, and the affinity KD value of the two recombinant antibodies is measured on a Biacore 8kplus instrument. The results are shown in FIG. 7 and Table 10.
Table 10
Immobilized ligands | Sample injection analyte solution | KD(M) | 1:1 Binding ka (1/Ms) | kd(1/s) |
MSLN | NM1 | 1.66E-09 | 5.52E+05 | 9.14E-04 |
MSLN | NM3 | 4.31E-11 | 1.31E+06 | 5.77E-05 |
MSLN | NM5 | 1.80E-10 | 6.08E+05 | 1.09E-04 |
MSLN | NM11 | 4.03E-10 | 2.26E+06 | 9.09E-04 |
MSLN | NM12 | 6.74E-10 | 6.04E+07 | 4.07E-02 |
MSLN | NM13 | 1.35E-10 | 5.80E+07 | 7.84E-03 |
MSLN | NM16 | 1.17E-09 | 6.58E+07 | 7.69E-04 |
MSLN | NM17 | 3.00E-09 | 2.15E+05 | 6.45E-04 |
MSLN | NM20 | 7.40E-10 | 4.05E+05 | 3.00E-04 |
MSLN | NM23 | 1.78E-09 | 3.34E+05 | 5.93E-04 |
MSLN | NM24 | 5.90E-10 | 9.59E+06 | 5.66E-03 |
Example 2: structural design of MSLN-targeted STAR
The variable regions (antigen binding regions) of the human TCR alpha and beta chains were replaced with alpaca-derived antibody heavy chain variable regions (VHH) capable of specifically recognizing Mesothelin (MSLN) and the following modifications were made in the constant regions of TCR alpha and beta chains: the human TCR alpha and beta chain sequences were replaced with murine sequences; two chains each introduced a cysteine mutation; introducing hydrophobic mutation into the transmembrane region of the alpha chain; deletion of two chains of intracellular amino acids; the intracellular connection of the two chains co-stimulatory factors. Thus, a mutant synthetic T cell receptor antigen receptor (SYNTHETIC T-Cell Receptor and AntigenReceptor, STAR) capable of specifically recognizing MSLN was constructed, the structure of which is shown in FIG. 2.
1. Design of wild type T cell receptor and constant region mutant STAR molecule thereof
1) Prototype design of STAR
The secreted antibodies (abs) or B Cell Receptors (BCR) produced by B cells have a great similarity to the T Cell Receptor (TCR) in terms of genetic structure, protein structure and spatial conformation. Both antibodies and TCRs are composed of variable and constant regions, with the variable region serving for antigen recognition and binding and the constant region serving for structural interactions and signal transduction. By replacing the variable regions of the TCR alpha and beta chains (or TCR gamma and delta chains) with the heavy chain variable region (VH) and the light chain variable region (VL) of the antibody, an artificially synthesized chimeric molecule, termed a synthetic T Cell Receptor antigen Receptor (SYNTHETIC T-Cell Receptor AND ANTIGEN Receptor, STAR/WT-STAR), can be constructed, the structure of which is shown in figure 2 (left).
The STAR molecule has two chains, the first chain being obtained by fusing the antigen recognition region to the constant region of the T cell receptor and the second chain being obtained by fusing the antigen recognition region to the constant region of the T cell receptor. The antigen recognition domain in the construct can be derived from antibody variable regions (such as VH, VL, scFv or VHH) of human, mouse, alpaca and rabbit, the T cell receptor constant region can be derived from constant regions of TCR alpha and beta chains or gamma and delta chains of human or mouse, and a plurality of constructs with different configurations and similar functions can be obtained through different combinations of the TCR constant regions of the antigen recognition regions.
After expression of both chains of STAR molecules in T cells, they bind to the CD3 epsilon delta, CD3 gamma epsilon, CD3 zeta chains endogenous to the cells in the endoplasmic reticulum to form complexes of 8 subunits and are displayed on the cell membrane surface as complexes. An intact STAR complex contains 10 immune receptor tyrosine activation motifs (Immunoreceptor Tyrosine-based Activation Motif, ITAM) (1 ITAM sequence in each of the intracellular domains of CD3 epsilon, delta, gamma, epsilon chains, 3 ITAM sequences in each of the intracellular domains of CD3 zeta chains) when the antigen recognition sequence of the STAR receptor binds to its specific antigen, the intracellular ITAM sequences are phosphorylated, thereby activating downstream signaling pathways, activating transcription factors such as NF-. Kappa.beta., NFAT, and AP-1, triggering T cell activation, and producing effector functions. Previous studies by the present inventors have shown that STAR is capable of mediating a stronger T cell activation signal than conventional chimeric antigen receptors (Chimericantigenreceptor, CAR), that STAR-T cells have significantly reduced background activation in the absence of antigen stimulation and are less prone to exhaustion with continued antigen stimulation, thus having significant advantages (see chinese patent application No. 201810898720.2). However, the present invention provides further improvements to STAR.
2) Design of mutant STAR (mut-STAR) and transmembrane and intracellular region modified STAR (ub-STAR)
STAR prototypes used constant region sequences for human TCR alpha and beta chains (or TCR gamma and delta chains) (wild type human TCR alpha constant region, SEQ ID NO:1; wild type human TCR beta constant region, SEQ ID NO: 2). The constant region sequences of human, primate and murine TCR alpha/beta chains can be interchanged due to their high functional conservation and identical key amino acid sequences.
After the STAR molecule is transduced into a T cell, a mismatch will occur with the TCR endogenous to the T cell via the constant region. This mismatch problem reduces the efficiency of correct pairing of STAR molecules, impairing their function, on the one hand, and increases the likelihood of mismatches generating unknown specific receptor molecules, increasing the safety risk, on the other hand. To solve this problem, the inventors have made the following modifications to the wt-STAR molecule: a. the constant region of the STAR molecule is replaced by a murine sequence, (wild-type murine TCR alpha constant region mouse TCRaC-WT, SEQ ID NO:3; wild-type murine TCR beta constant region mouse TCRbC-WT, SEQ ID NO: 4) so that the murine constant region cannot be mismatched with an endogenous human TCR chain to increase the efficiency of pairing between the two chains of the exogenous transferred STAR molecule. b. The STAR molecule constant region is subjected to cysteine point mutation, specifically, threonine T at position 48 is mutated into cysteine C (mouse TCRaC-Cys, SEQ ID NO: 5) in the TCR alpha chain constant region, and serine S at position 56 is mutated into cysteine C (mouse TCRbC-Cys, SEQ ID NO: 6) in the TCR beta chain constant region. These two newly added cysteines form disulfide bonds between the two chains of STAR, reduce mismatches between the two chains of STAR and the endogenous TCR chain, and help the STAR molecule form a more stable complex, thereby achieving better functions. In addition, to further increase the stability of STAR molecules and to increase the function of STAR-T cells, the inventors have also performed the following modifications to STAR molecules: c. hydrophobic amino acid substitutions are made in the transmembrane region of STAR, specifically 3 amino acid mutations in the 111 to 119 amino acid regions of the transmembrane region of the constant region of TCR alpha chain: serine S at position 112 is changed to leucine L, methionine M at position 114 is changed to isoleucine I, glycine G at position 115 is changed to valine V. The overall amino acid sequence of this region was changed from LSVMGLRIL to LLVIVLRIL, and this modification was designated mouse TCRaC-TM9, resulting in a constant region sequence of SEQ ID NO. 7. This design increases the hydrophobicity of the transmembrane region, counteracts the instability caused by the positive charge carried by the TCR transmembrane region, and allows the STAR molecule to exist more stably on the cell membrane, thus achieving better function, the structure of which is shown in figure 2 (middle). d. Mutation of lysine in the constant region of the alpha chain and the constant region of the beta chain of the STAR molecule into arginine produces Mouse TCR alpha C-Arg mut (SEQ ID NO: 8) and Mouse TCR beta C-Arg mut (SEQ ID NO: 9) as constant region sequences, respectively, which reduces the possibility of ubiquitination of the STAR molecule transmembrane region and intracellular end, thereby reducing endocytosis of the STAR molecule, enabling the STAR molecule to exist more stably on the cell membrane, and further obtaining better functions, the structure is shown in figure 2 (right).
2. Design of wild-type and mutant STAR molecules comprising co-stimulatory molecule receptor intracellular domains
In order to improve the proliferation capacity, survival time and the capability of infiltrating into tumor microenvironment of STAR-T cells in vivo and killing target cells with high efficiency, the inventor designs a novel structure, modifies STAR compound, and can customize enhanced mut-STAR cells according to requirements so as to improve the clinical response rate of STAR-T cells and realize durable curative effect.
1) Design of mut-STAR molecule (co-STAR) comprising co-stimulatory molecule receptor intracellular domains
In order to enhance STAR-T cytotoxicity and cell proliferation persistence, the present invention introduces human co-stimulatory molecule receptor intracellular end sequences into the C-terminus of the STAR constant region (FIG. 3), respectively, exploring their effect on STAR-T cell function. The STAR constant regions described herein include unmodified wt-STAR constant regions, cys-STAR constant regions containing additional intermolecular disulfide bonds, murine hm-STAR constant regions, and mut-STAR incorporating the three modifications described in section 1. The co-stimulatory signaling structure comprises an intracellular signaling domain of CD40, OX40, ICOS, CD28, 4-1BB or CD27, having the sequence SEQ ID NO 10,SEQ ID NO:11,SEQ ID NO:12,SEQ ID NO:13,SEQ ID NO:14,SEQ ID NO:15. The co-stimulatory molecule intracellular domains may be either alone or simultaneously in tandem at the C-terminus of the alpha chain of the TCR, or the C-terminus of the beta chain, or in parallel at the C-terminus of both the alpha and beta chains (co-STAR).
In addition, the co-stimulatory molecule domains may be linked directly to the C-terminus of the TCR constant region or through flexible linkers comprising glycine and serine (SEQ ID NO:60 to NO: 112), rigid linkers comprising alanine and glutamic acid (SEQ ID NO:49 to NO: 59), and cleavable-type linkers (SEQ ID NO:113 to NO: 117), or linked after removal of the intracellular end sequence of the TCR molecule (intracellular end removed and comprising cysteine-substituted and hydrophobic region engineered TCR alpha chain constant region (mouse TCRaC-del mut, SEQ ID NO: 16), intracellular end removed TCR beta chain constant region comprising cysteine-substituted (mouse TCR beta C-del mut, SEQ ID NO: 17)) (co-linker-STAR, FIG. 4). The multiple co-stimulatory molecule domains may also be linked directly or via a linker to the α and β chains or the C-terminus of the α or β chain of the TCR constant region (as shown by dCo-STAR or tCo-STAR, fig. 4) the TCR of γδ T cells consists of two chains, TCR γ and TCR δ, and γδ T cells may be divided into three subsets depending on the type of TCR δ chain, γδ1, γδ2 and γδ3, the different subsets being distributed differently in humans. γδ T cells recognize non-peptide antigens primarily in a non-MHC restricted manner, playing an important role in pathogen and tumor monitoring. The inventors introduced human co-stimulatory molecule receptor intracellular end sequences into the C-terminal ends of tcrγ and tcrδ, respectively, and expected to improve performance of γδ T cells.
2) Design of CD3 molecule (co-CD 3-STAR) comprising co-stimulatory molecule receptor intracellular domains
The CD3 subunits, including the gamma, delta, epsilon and zeta chains, form T cell receptor complexes with TCR molecules, transmitting signals from extracellular to intracellular, thereby regulating the state of the cell in response to stimuli. To design enhanced TCR T cells, the present inventors engineered the CD3 molecule to increase the tumor killing, proliferation and residence time of the T cells in vivo, the human co-stimulatory receptor intracellular domain was introduced into the C-terminus of the CD3 gamma chain (SEQ ID NO: 18), delta chain (SEQ ID NO: 19), epsilon chain (SEQ ID NO: 20) and zeta chain (SEQ ID NO: 21) (FIG. 5). Expression of the engineered CD3 molecule in mut-STAR-T cells is desirable to increase its function.
3) Design of STAR molecules (cytokine-STAR, CK-STAR) comprising cytokine receptor stimulation regions
Cytokines play an important role in the proliferation, anti-tumor, differentiation and other functions of T cells. Binding of different cytokines to their receptors transmits signals from the extracellular to the intracellular end, thereby regulating the state of the cell in response to stimulation.
In addition, studies have shown that downstream molecule STAT5 (SEQ ID NO: 25) is activated at the intracellular end of the IL-2 receptor by a cascade, thereby enhancing transcription of T cell proliferation-related molecules, enhancing the proliferative capacity of CAR-T cells. To design enhanced STAR-T cells, to improve tumor killing, proliferation and residence time of T cells in vivo, the present inventors engineered STAR molecules to connect human cytokine receptor intracellular signaling regions (e.g., IL-2. Beta. Receptor intracellular IL2Rb, SEQ ID NO:22; IL-7. Alpha. Receptor intracellular end, SEQ ID NO:23; IL-21 receptor intracellular end, SEQ ID NO:24, etc.) in tandem with the C-terminus of TCR. Alpha. Chain, or the C-terminus of the beta. Chain, or the C-terminus of the alpha and beta. Chain, or alternatively, to connect STAT5 activating module (SEQ ID NO: 25) to IL-2. Beta. Or IL-7. Alpha. Receptor intracellular region ends (IL-2RbQ,SEQID NO:26;IL-7RbQ,SEQ ID NO:27) via G4S (FIG. 6).
Example 3: NM 5/24-VHH-based anti-MSLN STAR molecular vector construction
1. Carrier source
Vectors used in the present invention, including viral vectors, plasmid vectors, etc., are purchased from or synthesized by commercial companies, and the full-length sequences of these vectors are obtained, with well-defined cleavage sites known.
2. Fragment Source
The STAR mentioned in the present invention includes the above-mentioned wt-STAR, mut-STAR, ub-STAR, co-linker-STAR, CK-STAR, co-CD3-STAR, etc. Gene fragments such as the constant region of TCR, the intracellular region of the co-stimulatory molecule receptor, the intracellular signal transduction region of the cytokine receptor, the tag sequence, linker (linker) and the like are all derived from commercial sources for synthesis in the present invention.
The STAR antigen binding region fragment used in this example was derived from VHH of antibody NM5 targeting MSLN (its amino acid sequence is shown as SEQ ID NO:28, its nucleotide sequence is shown as SEQ ID NO: 118) and/or VHH of antibody NM24 targeting MSLN (its amino acid sequence is shown as SEQ ID NO:29, its nucleotide sequence is shown as SEQ ID NO: 119), wherein the anti-MSLN NM5 VHH comprises heavy chain CDR1 shown as SEQ ID NO:34, heavy chain CDR2 shown as SEQ ID NO:35 and heavy chain CDR3 shown as SEQ ID NO:36, and the anti-MSLN 24VHH comprises heavy chain CDR1 shown as SEQ ID NO:37, heavy chain CDR2 shown as SEQ ID NO:38 and heavy chain CDR3 shown as SEQ ID NO: 39. VHH sequences were codon optimized and synthesized by commercial companies.
3. Vector construction
The lentiviral vector pCDH-IRES-RFP used in the invention is obtained by restriction endonuclease NotI/NheI to obtain a linear vector, the gene fragments are obtained by synthesis and PCR methods, and the linear vector and each gene fragment are connected by a homologous recombination method to obtain a complete vector.
4. Construct structure
Different combinations of constant, antigen binding and intracellular regions of STAR constructs can result in constructs of different configurations but all specifically targeting MSLN, see in particular table 11, where part of the receptor structure is shown in figure 8.
TABLE 11
For example, this example constructs NM5-TCR β -co-linker-STAR structure on the basis of pCDH-IRES-RFP vector. The antigen binding sequence used was anti-MSLN NM5 VHH (SEQ ID NO: 28). The first constant region mouse TCRaC-Cys-TM9 (SEQ ID NO: 31) and the second constant region mouseTCRbC-Cys (SEQ ID NO: 6) are used, the linker to which the co-stimulatory factor is attached is a flexible type linker (G4S) 3 (SEQ ID NO: 83), but may also be other flexible, rigid or cleavable type linkers (ranging from SEQ ID NO:49 to NO: 117). In addition, when the flexible linker is (G4S) 5 (SEQ ID NO: 85) and the C-terminal ends of the TCR alpha and beta chain constant regions are connected in parallel to two co-stimulatory factors, the anti-MSLN-STAR receptor structure also exhibits excellent membranous rate and killing effect.
5. Plasmid extraction
The basic lysis method is adopted for plasmid extraction, and the general principle is that bacterial suspension is exposed to strong anionic detergent with high pH, cell walls are broken, chromosome DNA and protein are denatured, the bacterial suspension is mutually entangled into large-scale complexes, the complexes are covered by dodecyl sulfate, when potassium ions are used for replacing sodium ions, the complexes effectively precipitate from the solution, and plasmid DNA can be recovered from supernatant after centrifugal removal. The plasmid concentration is usually detected by 260nm light absorption after plasmid extraction according to the bacterial liquid which is divided into small extract (2-5 mL), small extract medium amount (10-20 mL) and large extract (100-200 mL). The plasmid with correct primary sequencing result needs to be transformed to pick up a monoclonal, carry out small extraction and enzyme digestion to identify whether the plasmid is correct or not.
6. Packaging virus
Lentix-293T cells were inoculated into 10cm dishes at 5X 10 5/mL, incubated in a 5% CO 2 incubator at 37℃and transfected when cell density reached about 80% (observed under a microscope). Four plasmids were prepared according to PMD2.G: PRSV-Rev: PMDlg: TRANSFER PLAMID = 1:1:2:4 ratio, mixed homogeneously with 500 μl serum-free DMEM. mu.L of PEI-Max was mixed with 500. Mu.L of serum-free DMEM and allowed to stand at room temperature for 5min (PEI-Max to plasmid volume/mass ratio 3:1). The PEI-max mixture was slowly added to the plasmid mixture, gently swirled, mixed well and allowed to stand at room temperature for 15min. And slowly adding the final mixed solution into a culture medium, fully and uniformly mixing, putting the mixture back into an incubator for continuous culture for 12-16 h, changing the culture medium into a 6% FBS DMEM culture medium for continuous culture, and collecting the virus liquid for 48h and 72 h.
7. Virus titer measurement
The TCR knocked-out Jurkat-C4 cells were seeded at 1.5X10-5/mL in flat bottom 96-well plates, and 100. Mu.L of 1640 medium containing 10% FBS, 0.2. Mu.L of 1000 Xpolybrene was added to each well. The virus was diluted 10-fold in 1640 complete medium, and the amount of the first pore virus was 100. Mu.L in the case of the virus stock solution and 1. Mu.L in the case of the concentrate. The diluted cells were added to the virus wells, mixed at 32℃and 1500rpm, centrifuged for 90min, and cultured in a 5% CO 2 incubator at 37℃for 72 hours. Cells on a 96-well flat bottom plate were aspirated into a round bottom 96-well plate, centrifuged at 1800rpm at 4℃for 5min, and the supernatant was discarded. After 200. Mu.L of 1 XPBS was added, the mixture was centrifuged at 1800rpm at 4℃for 5min, and the supernatant was discarded. 200. Mu.L of 4% tissue fixative was added and stored in the dark and up-flow cytometer. Measuring infection efficiency by using a flow cytometer, selecting a hole with the infection rate of 2-30% when calculating titer, wherein the calculation formula is as follows: titer (TU/mL) =1.5X10≡4×positive rate ≡viral volume (. Mu.L) ×1000. The above viruses were infected with T cells and STAR was expressed.
Example 4: expression of STAR receptor and mutant thereof in T cells and detection of upper membrane
1. Isolation and activation of human primary T cells
After primary T cells were obtained by Ficoil isolation, they were cultured in X-VIVO medium containing 10% FBS and 100IU/mL IL-2 at an initial culture density of 1X 10 6/mL and activated by addition to CD3, CD28 and fibractin pre-coated well plates.
2. Infection of human primary T cells
After the primary T cells are activated for 24 hours, adding a virus liquid, centrifuging at 1500rpm for 90 minutes, and placing the mixture in a CO 2 incubator for culture. X-VIVO medium containing 10% FBS and 100IU/mL IL-2 was supplemented 24h after infection and plated out, and subcultured at 1-2 days later.
STAR upper Membrane detection
After 72 hours post infection, STAR upper membrane efficiency was measured by using Anti-mouse TCR β flow antibodies or MSLN protein labeled with a fluorescent tag (wherein the amino acid sequence of the red fluorescent protein is shown in SEQ ID NO: 30). Mismatch efficiency was assessed by comparing the RFP positive ratio to the positive ratio of Anti-mouse tcrp β flow antibody or fluorescent tagged MSLN protein staining. The results are shown in FIG. 9, where NM5, NM13, NM24STAR infection efficiencies are close based on RFP + subgroup ratio.
Example 5: in vitro killing ability and cytokine release level of anti-MSLN STAR comprehensive mutant based on NM5/NM24-VHH
As shown in FIGS. 10A and 10B, the present example selects pancreatic cancer tumor cell line (AsPC-1) and antigen-overexpressing cell lines (293T, 293T-hMSLN, and 293T-mMSLN) as target cells expressing MSLN.
In vitro co-culture of T cells and target cells
1) Comparison of killing of AsPC-1 cells by MSLN-STAR based on NM5 and NM24
Target cells were plated in 24 well plates overnight at a 1E 5/well density one day in advance for culture, and corresponding numbers of STAR-T cells were added to the target cells at a ratio of STAR positive T cells to target cells of 3:1, 1:1, 1:3, and the killing efficiency of the STAR-T cells on the target cells after 24 hours or 48 hours of culture was examined.
2) Comparison of killing efficiency of MSLN-STAR with different structures on AsPC-1 cells
MSLN-STAR-T cells of different structures shown in Table 11 were co-cultured with AsPC-1 target cells at an effective target ratio (E/T ratio) of 1:1 or 1:3, the co-cultivation time was 24 hours or 48 hours, and the killing efficiency was measured after co-cultivation.
3) Comparison of NM5/NM24 based MSLN-STAR with other antibody based MSLN-STAR cells
The experimental group NM5/NM24-MSLN-STAR antigen binding region was anti-MSLN-NM5/24VHH (SEQ ID NO:28; SEQ NO: 29). The antigen binding region of the control STAR was derived from three published antibodies against MSLN (SEQ ID NO:40,SEQ ID NO:41,SEQ NO:42, corresponding nucleotide sequence: 128,SEQ ID NO:129,SEQ NO:130, respectively) after construction of experimental and control STAR-T cells, respectively, according to 1:1 effective target ratio, NM5-STAR, NM24-STAR and three control group-STAR-T cells were co-cultured with target cells AsPC-1 or 293-T cells overexpressing MSLN for 48h, respectively, and killing efficiency was examined.
2. Detection method
1) And (3) killing efficiency detection: luciferase method
After co-cultivation, the supernatant was removed, 400. Mu.L of 1 Xcell lysate (promega) was added to each well, and the mixture was centrifuged at 12000rpm for 10min with shaking at room temperature, and the supernatant was removed. mu.L of the supernatant was added to a white 96-well plate, 2 multiplex wells were used per well, and 50. Mu.L of luciferase substrate (promega) was added per well. The chemiluminescent value was detected using a multifunctional microplate reader with a gain value fixed at 100. Cell killing calculation: killing efficiency = 100% - (effector cell-target cell pore value)/(control cell-target cell pore value).
2) Cytokine release level detection
After T cells are stimulated with target cells or antigens, T cells are collected, centrifuged, and the supernatant is taken. TNF-alpha, IFN-gamma, IL-2ELISA kits were used as Human IL-2 Uncoated ELISA, human TNF-alpha Uncoated ELISA, human IFN-gamma Uncoated ELISA (accession numbers 88-7025, 88-7346, 88-7316, respectively). The method comprises the following specific steps: 10 Xcoating Buffer was diluted to 1X with ddH 2 O, coated antibody (250X) was added, mixed well and then added to a 96-well plate (ELISA-specific) at 100. Mu.L/well. After sealing with the preservative film, the film was washed 3 times with 1 XPBST (also known as Wash Buffer,1 XPBS plus 0.05% Tween 20) at 260. Mu.L/well each time, diluted 1X with ddH 2 O5 XPELISA/ELISPOT reagent, added to 96-well plates at 200. Mu.L/well and allowed to stand at room temperature for 1h overnight. PBST was washed 1 time, standard curves were diluted (ranges: 2-250, 4-500, respectively), and samples were diluted 20-50 times using 1 XDiluent. Adding sample and standard curve, incubating for 2 hr at normal temperature in each well with 100 μl, washing with PBST three times, adding 1×Diluent diluted detection antibody, incubating for 1 hr,
PBST was washed 3 times, then 1 XDluent diluted HRP was added, incubated for 30 minutes, washed 6 times, TMB was added for development, development time was not more than 15 minutes, addition was stopped by 2N H 2SO 4, and light absorption was measured at 450 nm.
3. Test results
Co-culture results show that both NM5-STAR and NM24-STAR can effectively kill MSLN positive target cells AsPC-1 and 293T-hMSLN, but cannot recognize and kill 293T-mMSLN target cells, thus showing good specificity (see FIGS. 11A-D). And compared with different STAR structures, the STAR-T cell killing function of only one antibody VHH is better. Meanwhile, both NM5-STAR-T and NM24-STAR-T exhibited stronger killing ability and higher cytokine release level than STAR-T cells constructed with the reported antibodies (see FIGS. 12A-D).
Example 6: in vivo killing Activity of anti-MSLN STAR comprehensive mutant based on NM5/NM24VHH
1. Tumor model establishment
NSG immunodeficient mice were used in this experiment. The mouse genotype is NOD-Prkdcem IL2rg em26/Nju, lacks T cells, B cells and NK cells, and has defects in macrophages and dendritic cells. NSG mice are the most complete mouse strain with immunodeficiency at present, and can not generate rejection reaction to transplanted tumor and T cells, so that the NSG mice are widely applied to preclinical research of T cell treatment. Female NSG mice of 6-8 weeks of age were used in this experiment, and the weight difference of the mice in each experiment was controlled within 2 g. Mice were housed in separate ventilated cages within clean grade barriers free of Specific Pathogens (SPF), providing normal diet and pH biased drinking water to prevent pathogen contamination.
The experiment adopts human pancreatic cancer cell line AsPC-1 cells for xenogeneic subcutaneous or intraperitoneal inoculation transplantation. The AsPC-1 cells are cell lines expressing luciferase genes through lentiviral vectors, and the development and change of tumors are monitored in real time in mice in a manner of chemiluminescence through luciferin and in vivo imaging. In this model, different doses of AsPC-1-luciferase cells were inoculated into 6-8 week old female NSG mice by subcutaneous or intraperitoneal inoculation. Mice were intraperitoneally injected with a potassium salt solution of fluorescein every 3-4 days, and tumor cells were examined for fluorescent signals by in vivo imaging.
All manipulations of experimental mice were performed after passing the trial of experimental Animal study and use plan (Animal protocol).
2. Evaluation of xenograft tumor killing Activity by NM5/NM24 based STAR-T cells
Using the tumor model described above, asPC-1 cells were inoculated subcutaneously or intraperitoneally into mice at a dose of 5X 10 5/mouse, the mice were imaged in vivo after 3 days, the mice were grouped according to the size of the tumor fluorescent signal values, and after 1 day, control T cells or MSLN-STAR-T cells were reinfused by intravenous inoculation at a dose of 5X 10 6 RFP positive T cells/mouse, and the total reinfusion number of control T cells was kept consistent with that of STAR-T cell groups. After reinfusion, mice were subjected to fluorescent imaging every 3-7 days to detect tumor size. At the same time, the mouse body weight was measured to evaluate the potential toxicity of STAR-T cells to mice, and the results are shown in FIGS. 13A-F.
Example 7: effect of two or more co-stimulatory intracellular domains connected directly or via a linker in series to the C-terminus of the alpha and beta chains of a TCR or to the C-terminus of the alpha or beta chains on anti-MSLN STAR-T cell function
T cell and target cell in vitro co-culture method
For detection of anti-MSLN co-STAR T cell killing function, target cells are spread in 24-well plates for overnight culture according to the density of 1X 10 5/well one day in advance, and STAR positive T cells and target cells are added into the target cells for co-culture according to the ratio of STAR positive T cells to target cells of 3:1, 1:1 and 1:3, and different co-culture time groups are set, wherein the co-culture time groups are as follows: 24h,48h. For detection of proliferation of co-STAR T cells, target cells were incubated with primary T cells for 7 days to observe changes in cell proliferation number and IL-2 secretion, and positive T cells were obtained by flow cytometry after 7 days, were resting for two days in the absence of antigen stimulation, and were again incubated with target cells for 24h to detect T cell killing.
T cell stimulation by target antigen
The target antigen of the invention is generally cell surface protein, can be directly used for activating T cells to detect the functions of the T cells, is usually added with positive T cells of 1X 10 5/hole, is centrifuged, collects cell suspension or culture supernatant after 24h activation to detect the functions of the T cells, or is activated for 24h,48h,96h or 7 days to detect the killing functions of the T cells.
T cell killing efficiency detection: luciferase method
After co-cultivation, the supernatant was removed, 400. Mu.L of 1 Xcell lysate (Promega) was added to each well, and the mixture was centrifuged at 12000rpm for 10min with shaking at room temperature, and the supernatant was removed. mu.L of the supernatant was added to a white 96-well plate, 2 multiplex wells were used per well, and 50. Mu.L of luciferase substrate (Promega) was added per well. The chemiluminescent value was detected using a multifunctional microplate reader with a gain value fixed at 100. Cell killing calculation: killing efficiency = 100% - (effector cell-target cell pore value)/(control cell-target cell pore value).
And detecting the tumor killing effect of the anti-MSLN-co-STAR under the condition of different effective target ratios by using a luciferase detection method. Wherein the co-stimulatory intracellular domains are connected directly or via a linker in series to the C-terminus of the alpha and beta chains of the TCR or to the C-terminus of the alpha or beta chain, and wherein a plurality of co-stimulatory intracellular domains may be connected directly or via a linker (structure shown in FIGS. 4 and 8).
4.T cell secretion cytokine assay: ELISA (enzyme-Linked immuno sorbent assay)
A large amount of cytokines are released during T cell activation to help T cells kill target cells or promote expansion of T cells themselves, commonly referred to as TNF-alpha, IFN-gamma, IL-2. After T cells are stimulated with target cells or antigens, the T cells are collected and centrifuged, and the supernatant is taken. TNF- α, IFN- γ, IL-2ELISA kits were used at HumanIL-2UncoatedELISA, humanTNF- α UncoatedELISA, humanIFN- γ UncoatedELISA (88-7025, 88-7346, 88-7316, respectively). The method comprises the following specific steps: 10X CoatingBuffer was diluted to 1X with ddH 2 O, coated antibody (250X) was added, mixed well and then added to a 96-well plate (ELISA-specific), 100. Mu.L/well. After sealing with the preservative film, 1 XPBST (also known as WashBuffer,1 XPBS plus 0.05% Tween 20) was washed 3 times at 260. Mu.L/well, diluted 1 XELISA/ELISPOTDiluent with ddH 2 O, added to 96-well plates, 200. Mu.L/well and allowed to stand at room temperature for 1h overnight. PBST was washed 1 time, standard yeast dilutions (IL-2, TNF-alpha, IFN-gamma ranges 2-250, 4-500, respectively) were used, and samples were diluted 20-50 fold using 1 XDiluent. Adding a sample and standard yeast, incubating for 2h at normal temperature in 100 mu L of each well, washing with PBST three times, adding a1 xDiluent diluted detection antibody, incubating for 1h, washing with PBST 3 times, adding a1 xDiluent diluted HRP, incubating for 30 min, washing for 6 times, adding TMB for color development, keeping the color development time at no more than 15min, adding 2NH 2SO 4, and detecting light absorption at 450 nm.
5.T cell proliferation change detection: flow cytometer count
After 7 days of T cell co-incubation with target cells, centrifugation, PBS was resuspended to 200 μl and the change in positive T cell numbers was counted by flow cytometry. Change in T cell proliferation positive T cell number/initial positive T cell addition after fold proliferation = 7 days.
The separated anti-MSLN-co-linker-START cells and target cells were initially co-cultured according to an effective target ratio of 1:3, recorded as 0 days, and then the cells were collected for flow analysis at 1 day and 7 days, respectively. Wherein the culture medium is 1640 complete culture medium without IL2, the initial STAR-T cells are 1×10 5 cells, the samples at each time point are independently incubated, and the rest of the co-incubated samples are subjected to half-liquid exchange every other day, and the target cells are supplemented. Cells used for flow analysis are firstly stained with anti-humanCD antibody, and cells with specified volumes are collected and recorded when the system is on machine, and the number and the proportion of T cells in the system are obtained through conversion. The proliferation of T cells with co-stimulated intracellular domains connected in series at the C-terminal of the alpha chain or beta chain of TCR is analyzed by establishing a proliferation fold curve of the absolute anti-MSLN-co-linker-STAR cell number. And (3) analyzing the tumor clearance and proliferation capacities of STAR-T constructed by connecting two or more co-stimulatory molecule intracellular domains in series at the C end of the TCR alpha chain or the beta chain by combining a tumor killing effect result, an ELISA result and a T cell proliferation effect.
Example 8 Effect of Co-stimulatory intracellular domains containing different lengths of the G4S linker (linker) on STAR-T cell function by linking the alpha or beta constant region of the anti-MSLNSTAR receptor structure
T cell and target cell in vitro co-culture method
For detection of anti-MSLNco-START cell killing function, target cells are spread in 24 well plates for overnight culture according to the density of 1X 10 5/well one day in advance, and STAR-T cells with corresponding numbers are added into the target cells for co-culture according to the ratio of STAR positive T cells to target cells of 3:1, 1:1 and 1:3, and different co-culture time groups are set, wherein the co-culture time groups are as follows: 24h,48h.
2. Target antigen stimulates T cells
The target antigen of the invention is generally cell surface protein, can be directly used for activating T cells to detect the functions of the T cells, is usually added with positive T cells of 1X 10 5/hole, is centrifuged, collects cell suspension or culture supernatant after 24h activation to detect the functions of the T cells, or is activated for 24h,48h,96h or 7 days to detect the killing functions of the T cells.
T cell killing efficiency detection: luciferase method
After co-cultivation, the supernatant was removed, 400. Mu.L of 1 Xcell lysate (Promega) was added to each well, and the mixture was centrifuged at 12000rpm for 10min with shaking at room temperature, and the supernatant was removed. mu.L of the supernatant was added to a white 96-well plate, 2 multiplex wells were used per well, and 50. Mu.L of luciferase substrate (Promega) was added per well. The chemiluminescent value was detected using a multifunctional microplate reader with a gain value fixed at 100. Cell killing calculation: killing efficiency = 100% - (effector cell-target cell pore value)/(control cell-target cell pore value).
Detecting T cell killing function by luciferase method, wherein co-stimulated intracellular domain containing G4Slinker with different length is connected at the intracellular end of alpha or beta constant region of anti-MSLNSTAR acceptor structure, and the connection mode of alpha chain constant region is alpha-del-OX 40, alpha-del-G4S-OX 40, alpha-del- (G4S) 3-OX40, alpha-del- (G4S) 5-OX40, alpha-del- (G4S) 7-OX40 and alpha-del- (G4S) 10-OX40; the linker is connected to the intracellular end of the beta constant region in the manner of beta-del-OX 40, beta-OX 40, beta-del- (G4S) 3-OX40, beta-del- (G4S) 5-OX40, beta-del- (G4S) 7-OX40 and beta-del- (G4S) 10-OX40, wherein partial structures are shown in the figure 4.
4.T cell secretion cytokine assay: ELISA (enzyme-Linked immuno sorbent assay)
A large amount of cytokines are released during T cell activation to help T cells kill target cells or promote expansion of T cells themselves, commonly referred to as TNF-alpha, IFN-gamma, IL-2. After T cells are stimulated with target cells or antigens, T cells are collected, centrifuged, and the supernatant is taken. TNF-alpha, IFN-gamma, IL-2ELISA kits were used as Human IL-2Uncoated ELISA, human TNF-alpha Uncoated ELISA, human IFN-gamma Uncoated ELISA (accession numbers 88-7025, 88-7346, 88-7316, respectively). The method comprises the following specific steps: 10 Xcoating Buffer was diluted to 1X with ddH 2 O, coated antibody (250X) was added, mixed well and then added to a 96-well plate (ELISA-specific) at 100. Mu.L/well. After sealing with the preservative film, the film was washed 3 times with 1 XPBST (also known as Wash Buffer,1 XPBS plus 0.05% Tween 20) at 260. Mu.L/well each time, diluted 1X with ddH 2 O5 XPELISA/ELISPOT reagent, added to 96-well plates at 200. Mu.L/well and allowed to stand at room temperature for 1h overnight. PBST was washed 1 time, standard yeast dilutions (IL-2, TNF-alpha, IFN-gamma ranges 2-250, 4-500, respectively) were used, and samples were diluted 20-50 fold using 1 XDiluent. Adding a sample and standard yeast, incubating for 2h at normal temperature in 100 mu L of each well, washing with PBST three times, adding a1 xDiluent diluted detection antibody, incubating for 1h, washing with PBST 3 times, adding a1 xDiluent diluted HRP, incubating for 30 min, washing for 6 times, adding TMB for color development, keeping the color development time at no more than 15min, adding 2NH 2SO 4, and detecting light absorption at 450 nm.
5.T cell differentiation change detection: flow cytometer analysis
During T cell activation, a large amount of cytokines and other chemokines are released, and signals are transduced into the nucleus through cytokines or chemokine receptors, regulating the change in T cell differentiation. T cell differentiation is from primitive T cells (naive) to central memory T cells (Tcm) to effector memory T cells (Tem) and finally to effector T cells (Teff). While the proliferation and persistence of T cells in the body is affected by the number of T cells that differentiate into central memory T cells (Tcm) to effector memory T cell (Tem) cell types. Memory T cells are classified into stem cell-state T cells, central memory T cells, and effector memory T cells. The proportion of central memory T cell differentiation affects the sustained killing of T cells in vivo. The ratio of primitive T cells to effector T cells affects the killing effect and persistence of T cells on tumors in vivo. The differentiation of T cells was known by measuring the expression of CD45RA and CCR7 on the surface of T cells using a flow cytometer. After 7 days of co-incubation of T cells with target cells, the T cells were stained with anti-human-CD45RA-Percp-cy5.5 and anti-human-CCR7-APC flow antibody for 30 minutes, centrifuged, washed once with PBS, fixed with 4% paraformaldehyde solution, and examined for T cell differentiation by flow cytometry.
Differentiation of central memory T cells of anti-MSLNSTAR constructed by different lengths of G4S linker was examined by flow cytometry. And (3) combining tumor killing results, ELISA results and flow-type detection of cell differentiation, and analyzing tumor killing effects of anti-MSLNSTAR receptor structures deleted at intracellular ends of alpha or beta constant regions by different lengths (G4S) linker connection and differentiation conditions of memory cell groups of IL-2 secretion and T cells.
Example 9: in vivo killing Activity of anti-MSLN STAR comprehensive mutants based on G4S linkers of different lengths
In vivo killing activity of anti-MSLN STAR complex mutants without the G4S linker and with the (G4S) 5 linker were tested separately in the same manner as in example 6, and the results are shown in FIGS. 14A-F.
EXAMPLE 10 NM24 STAR-T cell pharmacokinetic analysis experiments
High doses of STAR-T cells were returned in tumor-bearing mice, approximately 3E8/kg of mouse body weight. The tissues of the mice were collected from the placebo mice, 4 hours, 1 day, 3 days, 7 days, 14 days and 21 days after T cell reinfusion, respectively. The tissue selected in this embodiment includes: blood, bone marrow, tumor, heart, liver, spleen, lung, kidney, brain, ovary. The distribution and metabolism of STAR-T cells in various tissues was examined using Q-PCR.
The results are shown in FIGS. 15A-B, where STAR copy number is increased in all tissues. Day 14 this visit point has the highest STAR copy number; tumor, spleen, lung copy number proliferate most rapidly in all tissues.
Comparative example 1 in vitro injury killing and IFN-gamma secretion of NM24 STAR-T cells comparative test experiment of TCR 2 TC-210, a similar cell product
In the same manner as in example 5, cells of NM24 STAR-T and the like cell product TCR 2 TC-210 were tested for in vitro killing and IFN-gamma secretion, respectively. As shown in FIGS. 16A-B, the in vitro killing of NM24 STAR-T cells and IFN-gamma secretion was superior to that of the same cell product TCR 2 TC-210.
Comparative example 2 in vivo efficacy and safety of NM24 STAR-T cells test experiment with similar cell products TCR 2 TC-210
In vivo efficacy and safety of NM24 STAR-T and cells of the same cell product TCR 2 TC-210, respectively, were tested in the same manner as in example 6. As shown in FIGS. 17A and 17B, NM24 STAR-T cells have better in vivo potency than the like cell product TCR 2 TC-210. As shown in fig. 17C, mice lost less weight after reinfusion of NM24 STAR-T cells compared to the like cell product TCR 2 TC-210; as shown in FIG. 17D, a total of 3 mice died on day 25 of the same cell product TCR 2 TC-210 after reinfusion. Therefore, NM24 STAR-T cells are safer than the same cell product TCR 2 TC-210.
The preferred embodiments of the present invention have been described in detail above, but the present invention is not limited to the specific details of the above embodiments, and various simple modifications can be made to the technical solution of the present invention within the scope of the technical concept of the present invention, and all the simple modifications belong to the protection scope of the present invention.
In addition, the specific features described in the above embodiments may be combined in any suitable manner, and in order to avoid unnecessary repetition, various possible combinations are not described further.
Claims (45)
- A synthetic T cell receptor antigen receptor, characterized by:The synthetic T cell receptor antigen receptor comprises an alpha chain and a beta chain, wherein the alpha chain comprises a first target binding region and a first constant region, and the beta chain comprises a second target binding region and a second constant region; or alternativelyIi) the synthetic T cell receptor antigen receptor comprises a gamma chain, a delta chain, wherein the gamma chain comprises a first target binding region and a first constant region, and the delta chain comprises a second target binding region and a second constant region;Wherein the first target binding region and the second target binding region comprise one or more antigen binding regions, the plurality of antigen binding regions being the same or different, the plurality of antigen binding regions being linked directly or through a linker;The antigen binding region in the first target binding region comprises a heavy chain variable region of an antibody that specifically binds mesothelin, and the antigen binding region in the second target binding region comprises a light chain variable region of an antibody that specifically binds mesothelin; or the antigen binding region in the first target binding region comprises a light chain variable region of an antibody that specifically binds mesothelin and the antigen binding region in the second target binding region comprises a heavy chain variable region of an antibody that specifically binds mesothelin.
- The synthetic T cell receptor antigen receptor of claim 1, wherein the antigen binding region in the first target binding region comprises a single chain antibody or a single domain antibody that specifically binds mesothelin; and/or the antigen binding region in the second target binding region comprises a single chain antibody or a single domain antibody that specifically binds mesothelin;Preferably, the single chain antibody comprises a heavy chain variable region and a light chain variable region linked directly or through a linker;preferably, the antigen binding region in the first target binding region and the antigen binding region in the second target binding region bind to different regions (e.g., different epitopes) of mesothelin.
- The synthetic T cell receptor antigen receptor of claim 1 or 2, wherein the antigen binding region in the first target binding region comprises one or more single domain antibodies and/or wherein the antigen binding region in the second target binding region comprises one or more single domain antibodies;preferably, the antigen binding region of the first target binding region comprises a plurality of single domain antibodies that are the same or different;Preferably, the antigen binding region of the second target binding region comprises a plurality of single domain antibodies that are the same or different;Further preferred, the plurality of single domain antibodies are linked directly or through a linker.
- The synthetic T cell receptor antigen receptor according to claim 3, wherein the single domain antibody comprises a heavy chain variable region comprising CDR1-3, wherein,I) CDR1 comprises SEQ ID NO:34, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:35, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:36, and a nucleotide sequence shown in seq id no;Or alternativelyIi) CDR1 comprises SEQ ID NO:37, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:38, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:39, and a polypeptide having the amino acid sequence shown in seq id no.
- The synthetic T cell receptor antigen receptor of claim 3, wherein: the single domain antibody comprises SEQ ID NO:28 or 29.
- The synthetic T cell receptor antigen receptor of claim 1 wherein the antigen receptor is a cell receptor,I) The alpha chain and/or the beta chain has attached at its C-terminal end at least one functional domain attached directly or through a linker to the C-terminal end of the alpha chain and/or the beta chain;Or alternativelyIi) the gamma and/or delta chain has attached at its C-terminal end at least one functional domain attached directly or via a linker to the C-terminal end of the gamma and/or delta chain.
- The synthetic T cell receptor antigen receptor of claim 1 wherein the antigen receptor is a cell receptor,I) The intracellular regions of the alpha and/or beta chains in the synthetic T cell receptor antigen receptor are deleted;Or alternativelyIi) the intracellular region of the gamma and/or delta chain in the synthetic T cell receptor antigen receptor is deleted.
- The synthetic T cell receptor antigen receptor of claim 7 wherein the antigen receptor is a cell receptor,I) The alpha chain and/or the beta chain has attached at its C-terminal end at least one functional domain attached directly or through a linker to the C-terminal end of the alpha chain and/or the beta chain;Or alternativelyIi) the gamma and/or delta chain has attached at its C-terminal end at least one functional domain attached directly or via a linker to the C-terminal end of the gamma and/or delta chain.
- The synthetic T cell receptor antigen receptor according to any one of claims 6 to 8, wherein,I) The C-terminus of the α -chain in the synthetic T-cell receptor antigen receptor is linked to 1,2,3,4,5, 6, 7, 8, 9, 10 or more functional domains, and/or the C-terminus of the β -chain in the synthetic T-cell receptor antigen receptor is linked to 1,2,3,4,5, 6, 7, 8, 9, 10 or more functional domains;Or alternativelyIi) 1,2,3,4,5, 6, 7, 8, 9, 10 or more functional domains are C-terminally linked to the gamma chain in the synthetic T cell receptor antigen receptor, and/or 1,2,3,4,5, 6, 7, 8, 9, 10 or more functional domains are C-terminally linked to the delta chain in the synthetic T cell receptor antigen receptor;wherein the functional domains are the same or different.
- The synthetic T cell receptor antigen receptor of any one of claims 6-8, wherein the functional domain is a costimulatory molecule or fragment thereof, a co-inhibitory molecule or fragment thereof, a cytokine receptor or fragment thereof, or an intracellular protein or fragment thereof;Preferably, the costimulatory molecule is selected from the group consisting of CD40, OX40, ICOS, CD28, 4-1BB or CD27;preferably, the co-inhibitory molecule is selected from the group consisting of TIM3, PD1, CTLA4, LAG3;Preferably, the cytokine receptor is selected from the group consisting of interleukin receptor, interferon receptor, tumor necrosis factor superfamily receptor, colony stimulating factor receptor, chemokine receptor, growth factor receptor, and other membrane proteins;preferably, the intracellular protein is a domain of a T cell regulator, such as NIK.
- The synthetic T cell receptor antigen receptor of any one of claims 1-10, wherein the linker is selected from the group consisting of a rigid linker, a flexible linker, a cleavable linker, and a nonsensical amino acid;Preferably, the amino acid sequence of the rigid linker is selected from the group consisting of SEQ ID NO:49-59, one or more than two;Preferably, the flexible linker is selected from glycine and/or serine rich peptide fragments; preferably, the flexible linker is selected from the group consisting of SEQ ID NO:60-112, one or more than two;Preferably, the cleavable linker is selected from the group consisting of SEQ ID NO: 113-117.
- The synthetic T cell receptor antigen receptor of any one of claims 1 to 11, wherein the first constant region is a TCR α chain constant region or a TCR γ chain constant region, preferably a modified TCR α chain constant region or a modified TCR γ chain constant region;Preferably, the TCR α chain constant region is selected from a human TCR α chain constant region or a murine (preferably mouse) TCR α chain constant region;preferably, the TCR γ chain constant region is selected from a human TCR γ chain constant region or a murine (preferably mouse) TCR γ chain constant region.
- The synthetic T cell receptor antigen receptor of claim 12, wherein: the modified TCR a chain constant region is derived from a murine (preferably mouse) TCR a chain constant region, which comprises one or more modifications, relative to a wild-type murine (preferably mouse) TCR a chain constant region, at positions 6, 13, 15-18, 48, 112, 114, 115, which modifications are mutations or deletions; or alternativelyThe modified TCR a chain constant region is derived from a murine (preferably mouse) TCR a chain constant region, which comprises one or more modifications, relative to a wild-type murine (preferably mouse) TCR a chain constant region, at positions 13, 36, 47, 53, 58, 78, 98, 122, which modifications are mutations or deletions.
- The synthetic T cell receptor antigen receptor of claim 12, wherein: the modified TCR a chain constant region is derived from a murine (preferably mouse) TCR a chain constant region that comprises an amino acid at position 48, e.g., threonine T, mutated to cysteine C, relative to a wild-type murine (preferably mouse) TCR a chain constant region;The modified TCR a chain constant region is derived from a murine (preferably mouse) TCR a chain constant region comprising an amino acid such as serine S at position 112, changed to leucine L, an amino acid such as methionine M at position 114, changed to isoleucine I, and/or an amino acid such as glycine G at position 115, changed to valine V, relative to a wild-type murine (preferably mouse) TCR a chain constant region;The modified TCR a chain constant region is derived from a murine (preferably mouse) TCR a chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted relative to a wild-type murine (preferably mouse) TCR a chain constant region;the modified TCR a chain constant region is derived from a murine (preferably mouse) TCR a chain constant region comprising the amino acid K at position 122 substituted with R relative to a wild-type murine (preferably mouse) TCR a chain constant region;The modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region comprising an amino acid such as threonine T at position 48 mutated to cysteine C, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V, relative to a wild-type murine (preferably mouse) TCR α chain constant region;The modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region which comprises an amino acid such as threonine T at position 48 mutated to cysteine C, with amino acid K at position 122 substituted with R, relative to a wild-type murine (preferably mouse) TCR α chain constant region;The modified TCR a chain constant region is derived from a murine (preferably mouse) TCR a chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, and an amino acid such as threonine T at position 48 mutated to cysteine C, relative to a wild-type murine (preferably mouse) TCR a chain constant region;The modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region comprising an amino acid such as threonine T at position 48 mutated to cysteine C, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V, and an amino acid K at position 122 replaced with R, relative to a wild-type murine (preferably mouse) TCR α chain constant region;The modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, an amino acid such as threonine T at position 48 mutated to cysteine C, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V, an amino acid K at position 122 substituted with R;The modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, an amino acid such as threonine T at position 48 mutated to cysteine C, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V;The modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, an amino acid such as threonine T at position 48 mutated to cysteine C, amino acid K at position 122 substituted with R, relative to a wild-type murine (preferably mouse) TCR α chain constant region;The modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V, relative to a wild-type murine (preferably mouse) TCR α chain constant region;The modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, and amino acid K at position 122 substituted with R, relative to a wild-type murine (preferably mouse) TCR α chain constant region; or alternativelyThe modified TCR α chain constant region is derived from a murine (preferably mouse) TCR α chain constant region comprising an amino acid such as E at position 6 substituted with D, K at position 13 substituted with R, and amino acids 15-18 deleted, an amino acid such as serine S at position 112 changed to leucine L, an amino acid such as methionine M at position 114 changed to isoleucine I, an amino acid such as glycine G at position 115 changed to valine V, and an amino acid K at position 122 changed to R, relative to a wild-type murine (preferably mouse) TCR α chain constant region.
- The synthetic T cell receptor antigen receptor of claim 12, wherein: the first constant region comprises SEQ ID NO: 1.3, 5, 7, 8, 16, 31, 32 or 43.
- The synthetic T cell receptor antigen receptor of any one of claims 1-15, wherein: the second constant region is a TCR β chain constant region or a TCR δ chain constant region, preferably a modified TCR β chain constant region or a modified TCR δ chain constant region;preferably, the TCR β chain constant region is selected from a human TCR β chain constant region or a murine (preferably mouse) TCR β chain constant region;Preferably, the TCR delta chain constant region is selected from a human TCR delta chain constant region or a murine (preferably mouse) TCR delta chain constant region.
- The synthetic T cell receptor antigen receptor of claim 16, wherein: the modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region comprising one or more modifications, relative to a wild-type murine (preferably mouse) TCR β chain constant region, at positions 3, 6, 9, 11, 12, 17, 21-25, 56, 150, 168 or 170, which modifications are mutations or deletions; or alternativelyThe modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region, which comprises one or more modifications, relative to a wild-type murine (preferably mouse) TCR β chain constant region, at positions 9, 17, 23, 25, 49, 63, 103, 110, 150, 168, 170, which modifications are mutations or deletions.
- The synthetic T cell receptor antigen receptor of claim 16, wherein: the modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region comprising an amino acid at position 56, e.g., serine S, mutated to cysteine C, relative to a wild-type murine (preferably mouse) TCR β chain constant region;The modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region comprising a substitution of lysine at position 150, 168 or 170 with arginine;The modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region comprising an amino acid such as R at position 3 substituted with K, an amino acid such as T at position 6 substituted with F, K at position 9 substituted with E, S at position 11 substituted with a, L at position 12 substituted with V, and amino acids at positions 17, 21-25 deleted, relative to a wild-type murine (preferably mouse) TCR β chain constant region;The modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region comprising an amino acid at position 56, e.g. serine S, mutated to cysteine C, and lysine at position 150, 168 or 170 substituted with arginine, relative to a wild-type murine (preferably mouse) TCR β chain constant region;The modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region comprising an amino acid such as R at position 3 substituted with K, an amino acid such as T at position 6 substituted with F, K at position 9 substituted with E, S at position 11 substituted with a, L at position 12 substituted with V, and amino acids 17, 21-25 deleted, and an amino acid such as serine S at position 56 mutated to cysteine C, relative to a wild-type murine (preferably mouse) TCR β chain constant region;The modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region comprising an amino acid such as R at position 3 substituted with K, an amino acid such as T at position 6 substituted with F, K at position 9 substituted with E, S at position 11 substituted with a, L at position 12 substituted with V, and amino acids 17, 21-25 deleted, an amino acid such as serine S at position 56 mutated to cysteine C, and lysine at position 150, 168 or 170 substituted with arginine, relative to a wild-type murine (preferably mouse) TCR β chain constant region; or alternativelyThe modified TCR β chain constant region is derived from a murine (preferably mouse) TCR β chain constant region comprising an amino acid such as R at position 3 substituted with K, an amino acid such as T at position 6 substituted with F, K at position 9 substituted with E, S at position 11 substituted with a, L at position 12 substituted with V, and amino acids 17, 21-25 deleted, and lysine at position 150, 168 or 170 substituted with arginine, relative to a wild-type murine (preferably mouse) TCR β chain constant region.
- The synthetic T cell receptor antigen receptor of claim 16, wherein: the second constant region comprises SEQ ID NO: 2. 4, 6, 9, 17, 33 or 44.
- The synthetic T cell receptor antigen receptor of claim 1 or 2, wherein: the first target binding region is linked directly to the first constant region or via a linker, and/or the second target binding region is linked directly to the second constant region or via a linker.
- The synthetic T cell receptor antigen receptor of claim 20, wherein: the linker is selected from a rigid linker, a flexible linker, a cleavable linker or a nonsensical amino acid;Preferably, the amino acid sequence of the rigid linker is selected from the group consisting of SEQ ID NO:49-59, one or more than two;Preferably, the flexible linker is selected from glycine and/or serine rich peptide fragments; preferably, the flexible linker is selected from the group consisting of SEQ ID NO:60-112, one or more than two;Preferably, the cleavable linker is selected from the group consisting of SEQ ID NO: 113-117.
- A synthetic T cell receptor antigen receptor complex, characterized by: the complex comprising the synthetic T cell receptor antigen receptor of any one of claims 1-21, and CD3 epsilon, CD3 gamma, CD3 delta, and CD3 zeta.
- The synthetic T cell receptor antigen receptor complex of claim 22, wherein: the CD3 epsilon, CD3 gamma, CD3 delta and/or CD3 zeta are of human origin;preferably, the CD3 epsilon comprises SEQ ID NO:20, and a polypeptide comprising the amino acid sequence shown in seq id no;preferably, the CD3 γ comprises SEQ ID NO:18, an amino acid sequence shown in seq id no;Preferably, the CD3 delta comprises SEQ ID NO:19, an amino acid sequence shown in seq id no;preferably, said CD3 ζ comprises SEQ ID NO:21, and a polypeptide comprising the amino acid sequence shown in seq id no.
- An antibody or antigen-binding fragment, characterized in that: the antibody or antigen binding fragment comprises a heavy chain variable region comprising CDR1-3 and/or a light chain variable region wherein,I) CDR1 comprises SEQ ID NO:34, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:35, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:36, and a nucleotide sequence shown in seq id no;Or alternativelyIi) CDR1 comprises SEQ ID NO:37, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:38, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:39, and a polypeptide having the amino acid sequence shown in seq id no.
- The antibody or antigen-binding fragment of claim 24, wherein: the antibody or antigen binding fragment is a single chain antibody or a single domain antibody.
- The antibody or antigen-binding fragment of claim 24, wherein: the antibody or antigen binding fragment comprises SEQ ID NO:28 or 29.
- An antigen receptor comprising a transmembrane region, an intracellular region and one or more identical or different extracellular binding domains, wherein the extracellular binding domain is an extracellular antigen binding domain;wherein the extracellular antigen-binding domain comprises CDR1-3, wherein,I) CDR1 comprises SEQ ID NO:34, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:35, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:36, and a nucleotide sequence shown in seq id no;Or alternativelyIi) CDR1 comprises SEQ ID NO:37, CDR2 comprises the amino acid sequence set forth in SEQ ID NO:38, said CDR3 comprises the amino acid sequence set forth in SEQ ID NO:39, and a polypeptide having the amino acid sequence shown in seq id no.
- The antigen receptor of claim 27, wherein the extracellular antigen-binding domain comprises an antibody or antigen-binding fragment of any one of claims 24-26.
- The antigen receptor of claim 27, wherein the antigen receptor is a STAR, TCR, or CAR;Preferably, the transmembrane region is derived from human CD8;preferably, the intracellular domain is derived from 4-1BB, CD28 or CD3 zeta.
- The antigen receptor of claim 27, wherein the transmembrane region is linked directly or via a linker to one or more extracellular antigen binding domains.
- The antigen receptor of claim 30, wherein the linker is selected from the group consisting of a rigid linker, a flexible linker, a cleavable linker, and a nonsensical amino acid;Preferably, the amino acid sequence of the rigid linker is selected from the group consisting of SEQ ID NO:49-59, one or more than two;Preferably, the flexible linker is selected from glycine and/or serine rich peptide fragments; preferably, the flexible linker is selected from the group consisting of SEQ ID NO:60-112, one or more than two;Preferably, the cleavable linker is selected from the group consisting of SEQ ID NO: 113-117.
- A nucleic acid, characterized in that: the nucleic acid encodes the synthetic T cell receptor antigen receptor of any one of claims 1-21, the synthetic T cell receptor antigen receptor complex of any one of claims 22-23, the antibody or antigen binding fragment of any one of claims 24-26, the antigen receptor of any one of claims 27-31.
- A carrier, characterized in that: the vector comprising the nucleic acid of claim 32.
- A host cell, characterized in that: the host cell comprises the nucleic acid of claim 32 or the vector of claim 33.
- An immune cell, characterized in that: the immune cell expresses the synthetic T cell receptor antigen receptor of any one of claims 1-21, the synthetic T cell receptor antigen receptor complex of any one of claims 22-23, the antibody or antigen binding fragment of any one of claims 24-26, the antigen receptor of any one of claims 27-31.
- The immune cell of claim 35, wherein: the immune cell contains one or more of the nucleic acid of claim 32.
- The immune cell of claim 35 or 36, wherein: the immune cells are selected from T cells, treg cells, macrophages, NK cells, NKT cells, peripheral blood mononuclear cells, TIL cells or Dendritic Cells (DC).
- The immune cell of claim 35 or 36, wherein the immune cell is derived from a T cell of the subject.
- A method for preparing an immune cell, comprising transfecting the nucleic acid sequence of claim 32 into an immune cell for expression.
- A method for preparing a recombinant T cell, comprising the steps of:1) Obtaining the nucleic acid of claim 32 from a positive T cell clone;2) Isolating and culturing primary T cells;3) Delivering the nucleic acid obtained in step 1) to the primary T cell of step 2) to obtain a recombinant T cell expressing the synthetic T cell receptor antigen receptor of any one of claims 1-21.
- A method for preparing a synthetic T cell receptor antigen receptor, comprising the steps of:(1) Obtaining the nucleic acid of claim 32 from a positive T cell clone;(2) Connecting the nucleic acid obtained in the step (1) to a vector skeleton to obtain an expression vector;(3) Transforming the expression vector obtained in the step (2) into a host cell, and then inducing the expression thereof;(4) Obtaining the synthetic T cell receptor antigen receptor.
- Use of a synthetic T cell receptor antigen receptor according to any one of claims 1 to 21, a synthetic T cell receptor antigen receptor complex according to any one of claims 22 to 23, an antibody or antigen binding fragment according to any one of claims 24 to 26, an antigen receptor according to any one of claims 27 to 31, a nucleic acid according to claim 32, an immune cell according to any one of claims 35 to 38 for the preparation of a product for the diagnosis or treatment of a tumor.
- The use according to claim 42, wherein: the tumors include lymphoma, non-small cell lung cancer, leukemia, ovarian cancer, nasopharyngeal cancer, breast cancer, endometrial cancer, colon cancer, rectal cancer, gastric cancer, bladder cancer, lung cancer, bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, liver and bile duct cancer, esophageal cancer, renal cancer, thyroid cancer, head and neck cancer, testicular cancer, glioblastoma, astrocytoma, melanoma, myelodysplastic syndrome, and sarcoma; wherein the leukemia is selected from acute lymphoblastic (lymphoblastic) leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, multiple myeloma, plasma cell leukemia, and chronic myelogenous leukemia; the lymphoma is selected from hodgkin's lymphoma and non-hodgkin's lymphoma, including B-cell lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone B-cell lymphoma, T-cell lymphoma, and waldenstrom's macroglobulinemia; the sarcoma is selected from osteosarcoma, ewing sarcoma, leiomyosarcoma, synovial sarcoma, soft tissue sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma, and chondrosarcoma.
- A pharmaceutical composition characterized by: the pharmaceutical composition comprises the synthetic T cell receptor antigen receptor of any one of claims 1-21, the synthetic T cell receptor antigen receptor complex of any one of claims 22-23, the antibody or antigen binding fragment of any one of claims 24-26, the antigen receptor of any one of claims 27-31, the nucleic acid of claim 32, the immune cell of any one of claims 35-38.
- A kit, characterized in that: the kit comprises the synthetic T cell receptor antigen receptor of any one of claims 1-21, the synthetic T cell receptor antigen receptor complex of any one of claims 22-23, the antibody or antigen binding fragment of any one of claims 24-26, the antigen receptor of any one of claims 27-31, the nucleic acid of claim 32, and the immune cell of any one of claims 35-38.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN2021113081469 | 2021-11-05 | ||
CN202111308146 | 2021-11-05 | ||
PCT/CN2022/130106 WO2023078431A1 (en) | 2021-11-05 | 2022-11-04 | Synthetic t cell receptor and antigen receptor specifically binding to mesothelin and use thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN118139897A true CN118139897A (en) | 2024-06-04 |
Family
ID=86240697
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202280071907.XA Pending CN118139897A (en) | 2021-11-05 | 2022-11-04 | Synthetic T cell receptor antigen receptor specifically combined with mesothelin and application thereof |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN118139897A (en) |
WO (1) | WO2023078431A1 (en) |
Family Cites Families (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
GB201607968D0 (en) * | 2016-05-06 | 2016-06-22 | Crescendo Biolog Ltd | Chimeric antigen receptor |
WO2018166589A1 (en) * | 2017-03-15 | 2018-09-20 | Biontech Cell & Gene Therapies Gmbh | Antigen receptors and uses thereof |
CN108715616A (en) * | 2018-04-27 | 2018-10-30 | 上海恒润达生生物科技有限公司 | The Chimeric antigen receptor method and purposes of targeting humanized mesothelin |
CN108864310A (en) * | 2018-07-31 | 2018-11-23 | 苏州茂行生物科技有限公司 | A kind of building and its application of the CAR-T cell for targeting mesothelin and carrying PD-Ll blocking agent |
CN110818802B (en) * | 2018-08-08 | 2022-02-08 | 华夏英泰(北京)生物技术有限公司 | Chimeric T cell receptor STAR and application thereof |
CN109111528B (en) * | 2018-09-20 | 2021-06-22 | 杭州普略生物科技有限公司 | Chimeric antigen receptor with MSLN as target point |
CN111499767B (en) * | 2020-06-15 | 2020-09-29 | 华夏英泰(北京)生物技术有限公司 | Synthetic T cell receptor antigen receptor complex and application thereof |
-
2022
- 2022-11-04 CN CN202280071907.XA patent/CN118139897A/en active Pending
- 2022-11-04 WO PCT/CN2022/130106 patent/WO2023078431A1/en active Application Filing
Also Published As
Publication number | Publication date |
---|---|
WO2023078431A1 (en) | 2023-05-11 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7093771B2 (en) | Programmed Death 1 Ligand 1 (PD-L1) -binding protein and its usage | |
CN109096396B (en) | anti-PD-L1 humanized nano antibody and application thereof | |
US20230212319A1 (en) | Novel antigen binding domains and synthetic antigen receptors incorporating the same | |
US10118964B2 (en) | Construction and application of bispecific antibody HER2xCD3 | |
JP2019083821A (en) | Tagged chimeric effector molecules and receptors thereof | |
TW202018083A (en) | Diverse antigen binding domains, novel platforms and other enhancements for cellular therapy | |
CN110582509A (en) | Treatment of cancer using chimeric T cell receptor proteins with multispecific properties | |
CN113039205A (en) | Antibodies targeting CLL1 and uses thereof | |
US11198737B2 (en) | T cell-antigen coupler with Y177 mutation and methods of uses thereof | |
AU2013305838A1 (en) | Method and compositions for cellular immunotherapy | |
WO2020135559A1 (en) | Cd30-binding moieties, chimeric antigen receptors, and uses thereof | |
JP7078237B1 (en) | Anti-TSPAN8-anti-CD3 bispecific antibody and anti-TSPAN8 antibody | |
WO2021048564A2 (en) | Antigen-binding domain | |
WO2021244626A1 (en) | Chimeric antigen receptor targeting cldn18.2 and use thereof | |
TW202039578A (en) | Cd3 binding molecules | |
CN117106099A (en) | anti-DLL 3 chimeric antigen receptor and application thereof | |
US20240252534A1 (en) | Antigen-Binding Domain | |
CN118139897A (en) | Synthetic T cell receptor antigen receptor specifically combined with mesothelin and application thereof | |
EP4188952A1 (en) | Anti-cd22 single domain antibodies and therapeutic constructs | |
WO2024140710A1 (en) | Synthetic t cell receptor antigen receptor specifically binding to lilrb4 and use thereof | |
WO2023088295A1 (en) | Multi-specific antibody and pharmaceutical use thereof | |
CN114539423B (en) | Trispecific fusion protein and application thereof | |
US12121542B2 (en) | BCMA chimeric antigen receptor based on single domain antibody and use thereof | |
US20220218746A1 (en) | Bcma chimeric antigen receptor based on single domain antibody and use thereof | |
WO2024076864A2 (en) | Anti-ror1 antibodies and uses thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication |