EP3864034A1 - Cellule - Google Patents
CelluleInfo
- Publication number
- EP3864034A1 EP3864034A1 EP19791327.0A EP19791327A EP3864034A1 EP 3864034 A1 EP3864034 A1 EP 3864034A1 EP 19791327 A EP19791327 A EP 19791327A EP 3864034 A1 EP3864034 A1 EP 3864034A1
- Authority
- EP
- European Patent Office
- Prior art keywords
- dnsmad
- cell
- cells
- domain
- chimeric
- 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.)
- Withdrawn
Links
- 210000004027 cell Anatomy 0.000 claims abstract description 173
- 108091008874 T cell receptors Proteins 0.000 claims abstract description 57
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 claims abstract description 54
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 claims abstract description 53
- 230000009261 transgenic effect Effects 0.000 claims abstract description 23
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 74
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 63
- 150000007523 nucleic acids Chemical class 0.000 claims description 62
- 229920001184 polypeptide Polymers 0.000 claims description 60
- 238000000034 method Methods 0.000 claims description 45
- 102000040430 polynucleotide Human genes 0.000 claims description 36
- 108091033319 polynucleotide Proteins 0.000 claims description 36
- 239000002157 polynucleotide Substances 0.000 claims description 36
- 239000013598 vector Substances 0.000 claims description 35
- 102000039446 nucleic acids Human genes 0.000 claims description 29
- 108020004707 nucleic acids Proteins 0.000 claims description 29
- 201000010099 disease Diseases 0.000 claims description 18
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 18
- 239000008194 pharmaceutical composition Substances 0.000 claims description 18
- 102100025748 Mothers against decapentaplegic homolog 3 Human genes 0.000 claims description 17
- 101710143111 Mothers against decapentaplegic homolog 3 Proteins 0.000 claims description 17
- 102100025751 Mothers against decapentaplegic homolog 2 Human genes 0.000 claims description 16
- 101710143123 Mothers against decapentaplegic homolog 2 Proteins 0.000 claims description 16
- CDKIEBFIMCSCBB-UHFFFAOYSA-N 1-(6,7-dimethoxy-3,4-dihydro-1h-isoquinolin-2-yl)-3-(1-methyl-2-phenylpyrrolo[2,3-b]pyridin-3-yl)prop-2-en-1-one;hydrochloride Chemical compound Cl.C1C=2C=C(OC)C(OC)=CC=2CCN1C(=O)C=CC(C1=CC=CN=C1N1C)=C1C1=CC=CC=C1 CDKIEBFIMCSCBB-UHFFFAOYSA-N 0.000 claims description 15
- 108010077850 Nuclear Localization Signals Proteins 0.000 claims description 14
- 230000004186 co-expression Effects 0.000 claims description 13
- 102100025725 Mothers against decapentaplegic homolog 4 Human genes 0.000 claims description 10
- 101710143112 Mothers against decapentaplegic homolog 4 Proteins 0.000 claims description 10
- 238000004519 manufacturing process Methods 0.000 claims description 5
- 239000003814 drug Substances 0.000 claims description 3
- 210000001744 T-lymphocyte Anatomy 0.000 description 44
- 235000001014 amino acid Nutrition 0.000 description 42
- 229940024606 amino acid Drugs 0.000 description 36
- 150000001413 amino acids Chemical class 0.000 description 36
- 230000027455 binding Effects 0.000 description 36
- 108090000623 proteins and genes Proteins 0.000 description 36
- 235000018102 proteins Nutrition 0.000 description 33
- 102000004169 proteins and genes Human genes 0.000 description 33
- 206010028980 Neoplasm Diseases 0.000 description 32
- 102000005962 receptors Human genes 0.000 description 30
- 108020003175 receptors Proteins 0.000 description 30
- 239000000427 antigen Substances 0.000 description 29
- 108091007433 antigens Proteins 0.000 description 27
- 102000036639 antigens Human genes 0.000 description 27
- 108091028043 Nucleic acid sequence Proteins 0.000 description 24
- 239000012642 immune effector Substances 0.000 description 23
- 229940121354 immunomodulator Drugs 0.000 description 23
- 238000003776 cleavage reaction Methods 0.000 description 22
- 230000007017 scission Effects 0.000 description 22
- 230000011664 signaling Effects 0.000 description 20
- 125000003275 alpha amino acid group Chemical group 0.000 description 14
- 201000011510 cancer Diseases 0.000 description 14
- 230000014509 gene expression Effects 0.000 description 14
- 238000006467 substitution reaction Methods 0.000 description 14
- 108020004414 DNA Proteins 0.000 description 13
- 230000000694 effects Effects 0.000 description 12
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 11
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 11
- 102000016715 Transforming Growth Factor beta Receptors Human genes 0.000 description 11
- 108010092867 Transforming Growth Factor beta Receptors Proteins 0.000 description 11
- 238000012217 deletion Methods 0.000 description 10
- 230000037430 deletion Effects 0.000 description 10
- 230000035772 mutation Effects 0.000 description 10
- 125000006850 spacer group Chemical group 0.000 description 10
- 238000010361 transduction Methods 0.000 description 10
- 230000026683 transduction Effects 0.000 description 10
- 230000004568 DNA-binding Effects 0.000 description 9
- 238000003501 co-culture Methods 0.000 description 9
- 210000000822 natural killer cell Anatomy 0.000 description 9
- 230000035755 proliferation Effects 0.000 description 9
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical group C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 9
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 8
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 8
- 239000012636 effector Substances 0.000 description 8
- 238000000684 flow cytometry Methods 0.000 description 8
- 230000006870 function Effects 0.000 description 8
- 230000004068 intracellular signaling Effects 0.000 description 8
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 8
- 101100454807 Caenorhabditis elegans lgg-1 gene Proteins 0.000 description 7
- 108010076504 Protein Sorting Signals Proteins 0.000 description 7
- 230000004913 activation Effects 0.000 description 7
- 210000000130 stem cell Anatomy 0.000 description 7
- 238000001890 transfection Methods 0.000 description 7
- 108091027981 Response element Proteins 0.000 description 6
- 102000007374 Smad Proteins Human genes 0.000 description 6
- 108010007945 Smad Proteins Proteins 0.000 description 6
- 238000003556 assay Methods 0.000 description 6
- 239000011230 binding agent Substances 0.000 description 6
- 210000002865 immune cell Anatomy 0.000 description 6
- 239000003446 ligand Substances 0.000 description 6
- 230000001404 mediated effect Effects 0.000 description 6
- 125000003729 nucleotide group Chemical group 0.000 description 6
- 230000002829 reductive effect Effects 0.000 description 6
- 125000003607 serino group Chemical group [H]N([H])[C@]([H])(C(=O)[*])C(O[H])([H])[H] 0.000 description 6
- 230000002103 transcriptional effect Effects 0.000 description 6
- 102000004127 Cytokines Human genes 0.000 description 5
- 108090000695 Cytokines Proteins 0.000 description 5
- 102000004961 Furin Human genes 0.000 description 5
- 108090001126 Furin Proteins 0.000 description 5
- 101000798109 Homo sapiens Melanotransferrin Proteins 0.000 description 5
- 102100032239 Melanotransferrin Human genes 0.000 description 5
- 239000003795 chemical substances by application Substances 0.000 description 5
- 230000001461 cytolytic effect Effects 0.000 description 5
- 238000002784 cytotoxicity assay Methods 0.000 description 5
- 231100000263 cytotoxicity test Toxicity 0.000 description 5
- 230000003834 intracellular effect Effects 0.000 description 5
- 230000004048 modification Effects 0.000 description 5
- 238000012986 modification Methods 0.000 description 5
- 239000002773 nucleotide Substances 0.000 description 5
- 210000005259 peripheral blood Anatomy 0.000 description 5
- 239000011886 peripheral blood Substances 0.000 description 5
- 238000001262 western blot Methods 0.000 description 5
- 241000710198 Foot-and-mouth disease virus Species 0.000 description 4
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 4
- 206010035226 Plasma cell myeloma Diseases 0.000 description 4
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 4
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 4
- 241000723792 Tobacco etch virus Species 0.000 description 4
- 108010009583 Transforming Growth Factors Proteins 0.000 description 4
- 102000009618 Transforming Growth Factors Human genes 0.000 description 4
- 210000003719 b-lymphocyte Anatomy 0.000 description 4
- 210000004899 c-terminal region Anatomy 0.000 description 4
- 231100000135 cytotoxicity Toxicity 0.000 description 4
- 230000003013 cytotoxicity Effects 0.000 description 4
- 238000010586 diagram Methods 0.000 description 4
- 239000012634 fragment Substances 0.000 description 4
- 238000009169 immunotherapy Methods 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 230000002401 inhibitory effect Effects 0.000 description 4
- 230000003993 interaction Effects 0.000 description 4
- 230000002147 killing effect Effects 0.000 description 4
- 210000004698 lymphocyte Anatomy 0.000 description 4
- 210000004962 mammalian cell Anatomy 0.000 description 4
- 230000007246 mechanism Effects 0.000 description 4
- 230000026731 phosphorylation Effects 0.000 description 4
- 238000006366 phosphorylation reaction Methods 0.000 description 4
- 230000001177 retroviral effect Effects 0.000 description 4
- 230000019491 signal transduction Effects 0.000 description 4
- 230000004083 survival effect Effects 0.000 description 4
- 230000005945 translocation Effects 0.000 description 4
- 239000013603 viral vector Substances 0.000 description 4
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 3
- 241000710190 Cardiovirus Species 0.000 description 3
- 101000581981 Homo sapiens Neural cell adhesion molecule 1 Proteins 0.000 description 3
- 101001059454 Homo sapiens Serine/threonine-protein kinase MARK2 Proteins 0.000 description 3
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 3
- 208000034578 Multiple myelomas Diseases 0.000 description 3
- 102100028904 Serine/threonine-protein kinase MARK2 Human genes 0.000 description 3
- 108010076818 TEV protease Proteins 0.000 description 3
- 235000004279 alanine Nutrition 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 230000022534 cell killing Effects 0.000 description 3
- 238000000749 co-immunoprecipitation Methods 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 210000004700 fetal blood Anatomy 0.000 description 3
- 210000004475 gamma-delta t lymphocyte Anatomy 0.000 description 3
- 238000000670 ligand binding assay Methods 0.000 description 3
- 125000003588 lysine group Chemical class [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 3
- 210000002540 macrophage Anatomy 0.000 description 3
- 230000031852 maintenance of location in cell Effects 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 239000002243 precursor Substances 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 238000009877 rendering Methods 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 230000009258 tissue cross reactivity Effects 0.000 description 3
- YXHLJMWYDTXDHS-IRFLANFNSA-N 7-aminoactinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=C(N)C=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 YXHLJMWYDTXDHS-IRFLANFNSA-N 0.000 description 2
- 108700012813 7-aminoactinomycin D Proteins 0.000 description 2
- RZVAJINKPMORJF-UHFFFAOYSA-N Acetaminophen Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 2
- 108010052946 Activin Receptors Proteins 0.000 description 2
- 102000018918 Activin Receptors Human genes 0.000 description 2
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 2
- 108010077544 Chromatin Proteins 0.000 description 2
- 102100032174 GTP-binding protein SAR1a Human genes 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical group NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 239000004471 Glycine Substances 0.000 description 2
- 101000964562 Homo sapiens Zinc finger FYVE domain-containing protein 9 Proteins 0.000 description 2
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 2
- 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
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 description 2
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 2
- 102100027347 Neural cell adhesion molecule 1 Human genes 0.000 description 2
- 206010029260 Neuroblastoma Diseases 0.000 description 2
- 102000035195 Peptidases Human genes 0.000 description 2
- 108091005804 Peptidases Proteins 0.000 description 2
- 108091000080 Phosphotransferase Proteins 0.000 description 2
- 102000006437 Proprotein Convertases Human genes 0.000 description 2
- 108010044159 Proprotein Convertases Proteins 0.000 description 2
- 206010060862 Prostate cancer Diseases 0.000 description 2
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 2
- 230000006052 T cell proliferation Effects 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- DZBUGLKDJFMEHC-UHFFFAOYSA-N acridine Chemical group C1=CC=CC2=CC3=CC=CC=C3N=C21 DZBUGLKDJFMEHC-UHFFFAOYSA-N 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 2
- 210000002203 alpha-beta t lymphocyte Anatomy 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 235000009697 arginine Nutrition 0.000 description 2
- 230000003190 augmentative effect Effects 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 210000003995 blood forming stem cell Anatomy 0.000 description 2
- 210000001185 bone marrow Anatomy 0.000 description 2
- 210000003855 cell nucleus Anatomy 0.000 description 2
- 238000001516 cell proliferation assay Methods 0.000 description 2
- 230000005754 cellular signaling Effects 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 210000003483 chromatin Anatomy 0.000 description 2
- 238000004132 cross linking Methods 0.000 description 2
- 230000001086 cytosolic effect Effects 0.000 description 2
- 206010012818 diffuse large B-cell lymphoma Diseases 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 230000007783 downstream signaling Effects 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 230000007717 exclusion Effects 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 229950004003 fresolimumab Drugs 0.000 description 2
- 108020001507 fusion proteins Proteins 0.000 description 2
- 102000037865 fusion proteins Human genes 0.000 description 2
- 230000003394 haemopoietic effect Effects 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 210000000987 immune system Anatomy 0.000 description 2
- 230000008676 import Effects 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 238000001802 infusion Methods 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 235000018977 lysine Nutrition 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 201000001441 melanoma Diseases 0.000 description 2
- 239000000203 mixture Substances 0.000 description 2
- 239000003607 modifier Substances 0.000 description 2
- 238000003032 molecular docking Methods 0.000 description 2
- 210000004940 nucleus Anatomy 0.000 description 2
- 210000002568 pbsc Anatomy 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 102000020233 phosphotransferase Human genes 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 230000002062 proliferating effect Effects 0.000 description 2
- 230000006916 protein interaction Effects 0.000 description 2
- 230000002285 radioactive effect Effects 0.000 description 2
- 230000007115 recruitment Effects 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 150000003355 serines Chemical class 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 108010087967 type I signal peptidase Proteins 0.000 description 2
- 235000002374 tyrosine Nutrition 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
- 102100031585 ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Human genes 0.000 description 1
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 108010043324 Amyloid Precursor Protein Secretases Proteins 0.000 description 1
- 102000002659 Amyloid Precursor Protein Secretases Human genes 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 208000032116 Autoimmune Experimental Encephalomyelitis Diseases 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 108091008875 B cell receptors Proteins 0.000 description 1
- 102100023995 Beta-nerve growth factor Human genes 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 101000724917 Calliophis bivirgatus Delta-elapitoxin-Cb1a Proteins 0.000 description 1
- 101000724912 Calliophis bivirgatus Maticotoxin A Proteins 0.000 description 1
- 108090000227 Chymases Proteins 0.000 description 1
- 102000003858 Chymases Human genes 0.000 description 1
- 108020004705 Codon Proteins 0.000 description 1
- 108700010070 Codon Usage Proteins 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 102000005927 Cysteine Proteases Human genes 0.000 description 1
- 108010005843 Cysteine Proteases Proteins 0.000 description 1
- 101000724921 Dendroaspis polylepis polylepis Dendroaspis polylepis MT9 Proteins 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 206010014759 Endometrial neoplasm Diseases 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000214054 Equine rhinitis A virus Species 0.000 description 1
- 108010087819 Fc receptors Proteins 0.000 description 1
- 102000009109 Fc receptors Human genes 0.000 description 1
- 206010016654 Fibrosis Diseases 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
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 1
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000777636 Homo sapiens ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Proteins 0.000 description 1
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 1
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 1
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 1
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 1
- 101000712674 Homo sapiens TGF-beta receptor type-1 Proteins 0.000 description 1
- 101000712669 Homo sapiens TGF-beta receptor type-2 Proteins 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- IVRXNBXKWIJUQB-UHFFFAOYSA-N LY-2157299 Chemical group CC1=CC=CC(C=2C(=C3CCCN3N=2)C=2C3=CC(=CC=C3N=CC=2)C(N)=O)=N1 IVRXNBXKWIJUQB-UHFFFAOYSA-N 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 102100026964 M1-specific T cell receptor beta chain Human genes 0.000 description 1
- 102000043129 MHC class I family Human genes 0.000 description 1
- 108091054437 MHC class I family Proteins 0.000 description 1
- 102000011716 Matrix Metalloproteinase 14 Human genes 0.000 description 1
- 108010076557 Matrix Metalloproteinase 14 Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 206010027480 Metastatic malignant melanoma Diseases 0.000 description 1
- 101000783591 Micrurus clarki Clarkitoxin-1 Proteins 0.000 description 1
- 101000963932 Micrurus frontalis Frontoxin II Proteins 0.000 description 1
- 101000783588 Micrurus mipartitus Mipartoxin-1 Proteins 0.000 description 1
- 101000963935 Micrurus nigrocinctus Nicotinic acetylcholine receptor-binding protein Mnn-1A Proteins 0.000 description 1
- 101000964147 Micrurus nigrocinctus Nicotinic acetylcholine receptor-binding protein Mnn-3C Proteins 0.000 description 1
- 101000964140 Micrurus nigrocinctus Nicotinic acetylcholine receptor-binding protein Mnn-4 Proteins 0.000 description 1
- 101000724922 Micrurus pyrrhocryptus Venom protein E2 Proteins 0.000 description 1
- 101000724923 Micrurus surinamensis Short neurotoxin MS11 Proteins 0.000 description 1
- 102100030608 Mothers against decapentaplegic homolog 7 Human genes 0.000 description 1
- 125000000729 N-terminal amino-acid group Chemical group 0.000 description 1
- 101000724924 Naja kaouthia Nakoroxin Proteins 0.000 description 1
- 108010025020 Nerve Growth Factor Proteins 0.000 description 1
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 description 1
- 102000017954 Nuclear factor of activated T cells (NFAT) Human genes 0.000 description 1
- 108050007058 Nuclear factor of activated T cells (NFAT) Proteins 0.000 description 1
- 108020005497 Nuclear hormone receptor Proteins 0.000 description 1
- 102000007399 Nuclear hormone receptor Human genes 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 101000963934 Ophiophagus hannah Neurotoxin Oh9-1 Proteins 0.000 description 1
- 101000724920 Ophiophagus hannah Short neurotoxin OH-26 Proteins 0.000 description 1
- 101000963927 Ophiophagus hannah Short neurotoxin OH-32 Proteins 0.000 description 1
- 101000724910 Ophiophagus hannah Short neurotoxin OH-46 Proteins 0.000 description 1
- 101000964138 Ophiophagus hannah Short neurotoxin OH-5 Proteins 0.000 description 1
- 101000724915 Ophiophagus hannah Short neurotoxin SNTX11 Proteins 0.000 description 1
- 101000724916 Ophiophagus hannah Short neurotoxin SNTX14 Proteins 0.000 description 1
- 101000724918 Ophiophagus hannah Short neurotoxin SNTX26 Proteins 0.000 description 1
- 101000724908 Ophiophagus hannah Short neurotoxin SNTX6 Proteins 0.000 description 1
- 101000964146 Ophiophagus hannah Weak neurotoxin WNTX33 Proteins 0.000 description 1
- 101000964133 Oxyuranus microlepidotus Toxin 3FTx-Oxy5 Proteins 0.000 description 1
- 101000724919 Oxyuranus scutellatus scutellatus Scutelatoxin Proteins 0.000 description 1
- 101000964145 Oxyuranus scutellatus scutellatus Short neurotoxin 3 Proteins 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 241000709664 Picornaviridae Species 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 108010039918 Polylysine Chemical group 0.000 description 1
- 102000007584 Prealbumin Human genes 0.000 description 1
- 108010071690 Prealbumin Proteins 0.000 description 1
- 101710118538 Protease Proteins 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 101800001065 Protein 2B Proteins 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 102000005601 Receptor-Regulated Smad Proteins Human genes 0.000 description 1
- 108010084447 Receptor-Regulated Smad Proteins Proteins 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 108091081062 Repeated sequence (DNA) Proteins 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- 241000702670 Rotavirus Species 0.000 description 1
- 101700026522 SMAD7 Proteins 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temozolomide Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical group OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 208000024770 Thyroid neoplasm Diseases 0.000 description 1
- 102000046299 Transforming Growth Factor beta1 Human genes 0.000 description 1
- 101800002279 Transforming growth factor beta-1 Proteins 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- 241000223104 Trypanosoma Species 0.000 description 1
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 1
- 108091005906 Type I transmembrane proteins Proteins 0.000 description 1
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- FBCSDEQVNNRGEQ-DKWTVANSSA-N [P].OC[C@H](N)C(O)=O Chemical group [P].OC[C@H](N)C(O)=O FBCSDEQVNNRGEQ-DKWTVANSSA-N 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 229960005305 adenosine Drugs 0.000 description 1
- 229940100198 alkylating agent Drugs 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 150000001484 arginines Chemical class 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 230000020411 cell activation Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000030570 cellular localization Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 230000002860 competitive effect Effects 0.000 description 1
- 238000004624 confocal microscopy Methods 0.000 description 1
- 108091036078 conserved sequence Proteins 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 210000004405 cytokine-induced killer cell Anatomy 0.000 description 1
- 210000005220 cytoplasmic tail Anatomy 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 238000002337 electrophoretic mobility shift assay Methods 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 208000012997 experimental autoimmune encephalomyelitis Diseases 0.000 description 1
- 230000002349 favourable effect Effects 0.000 description 1
- 230000004761 fibrosis Effects 0.000 description 1
- 238000002875 fluorescence polarization Methods 0.000 description 1
- 238000002866 fluorescence resonance energy transfer Methods 0.000 description 1
- 238000005194 fractionation Methods 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 229950000456 galunisertib Drugs 0.000 description 1
- 238000012239 gene modification Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 230000005017 genetic modification Effects 0.000 description 1
- 235000013617 genetically modified food Nutrition 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 210000002288 golgi apparatus Anatomy 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 208000029824 high grade glioma Diseases 0.000 description 1
- 235000014304 histidine Nutrition 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 102000049018 human NCAM1 Human genes 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 230000008073 immune recognition Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 210000005007 innate immune system Anatomy 0.000 description 1
- 210000004964 innate lymphoid cell Anatomy 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- 238000000111 isothermal titration calorimetry Methods 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 238000000021 kinase assay Methods 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 229960002247 lomustine Drugs 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 210000003738 lymphoid progenitor cell Anatomy 0.000 description 1
- 230000002101 lytic effect Effects 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 201000011614 malignant glioma Diseases 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 208000021039 metastatic melanoma Diseases 0.000 description 1
- YACKEPLHDIMKIO-UHFFFAOYSA-N methylphosphonic acid Chemical compound CP(O)(O)=O YACKEPLHDIMKIO-UHFFFAOYSA-N 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 229940053128 nerve growth factor Drugs 0.000 description 1
- 230000003472 neutralizing effect Effects 0.000 description 1
- 230000012223 nuclear import Effects 0.000 description 1
- 230000025308 nuclear transport Effects 0.000 description 1
- 210000003463 organelle Anatomy 0.000 description 1
- 210000002741 palatine tonsil Anatomy 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- DCWXELXMIBXGTH-UHFFFAOYSA-N phosphotyrosine Chemical compound OC(=O)C(N)CC1=CC=C(OP(O)(O)=O)C=C1 DCWXELXMIBXGTH-UHFFFAOYSA-N 0.000 description 1
- 210000002826 placenta Anatomy 0.000 description 1
- 210000004180 plasmocyte Anatomy 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920000656 polylysine Chemical group 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 125000001500 prolyl group Chemical group [H]N1C([H])(C(=O)[*])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 230000001902 propagating effect Effects 0.000 description 1
- 108010022328 proparathormone Proteins 0.000 description 1
- 235000019833 protease Nutrition 0.000 description 1
- 108020001580 protein domains Proteins 0.000 description 1
- 230000004850 protein–protein interaction Effects 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- 230000014493 regulation of gene expression Effects 0.000 description 1
- 210000003289 regulatory T cell Anatomy 0.000 description 1
- 238000002271 resection Methods 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 230000008054 signal transmission Effects 0.000 description 1
- 238000009097 single-agent therapy Methods 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 210000002536 stromal cell Anatomy 0.000 description 1
- 230000004960 subcellular localization Effects 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 229960004964 temozolomide Drugs 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 235000008521 threonine Nutrition 0.000 description 1
- 210000001541 thymus gland Anatomy 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 108091006106 transcriptional activators Proteins 0.000 description 1
- 108091008023 transcriptional regulators Proteins 0.000 description 1
- 108091006107 transcriptional repressors Proteins 0.000 description 1
- 230000002463 transducing effect Effects 0.000 description 1
- 238000003151 transfection method Methods 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 229940099456 transforming growth factor beta 1 Drugs 0.000 description 1
- 238000003146 transient transfection Methods 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 1
- 125000001493 tyrosinyl group Chemical group [H]OC1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 108010047303 von Willebrand Factor Proteins 0.000 description 1
- 102100036537 von Willebrand factor Human genes 0.000 description 1
- 229960001134 von willebrand factor Drugs 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/463—Cellular immunotherapy characterised by recombinant expression
- A61K39/4631—Chimeric Antigen Receptors [CAR]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/464469—Tumor associated carbohydrates
- A61K39/464471—Gangliosides, e.g. GM2, GD2 or GD3
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70521—CD28, CD152
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/50—Cell markers; Cell surface determinants
- C12N2501/515—CD3, T-cell receptor complex
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2510/00—Genetically modified cells
Definitions
- the present invention relates to a cell which expresses a chimeric antigen receptor (CAR) or a transgenic T-cell receptor (TCR).
- CAR chimeric antigen receptor
- TCR transgenic T-cell receptor
- the cells may be engineered to be resistant to the immunosuppressive effects of cytokines such as TQRb which signal through the Smad family of transcription factors.
- Adoptive immunotherapy of cancer involves the ex vivo generation of cancer-antigen specific cells and their administration.
- Adoptively transferred immune effector cells also activate existing adaptive and innate immune cells within the tumour once they activate and start causing inflammation.
- the native specificity of immune effector cells can be exploited in adoptive immunotherapy - for example during the generation of melanoma specific T-cells from expansion of tumour infiltrating lymphocytes in tumour resections. Otherwise a specificity can be grafted onto a T- cell using genetic engineering. Two common methods for achieving this are using chimeric antigen receptors or transgenic T-cell receptors. Different kinds of immune effector cells can also be used. For example, alpha/beta T-cells, NK cells, gamma delta T-cells or macrophages can be used.
- B-ALL B-cell Acute Lymphoblastic Leukaemia
- DLBCL Diffuse Large B-cell Lymphoma
- MM Multiple Myeloma
- tumour microenvironment may convert the microenvironment into a more favourable environment which enables the engineered immune effector cells to proliferate, survive and/or engraft thereby providing a more effective engineered cell therapy.
- TQRb transforming growth factor beta
- Fresolimumab is a neutralizing antibody which blocks TQRb1-3.
- Fresolimumab has been tested in metastatic melanoma and high-grade glioma. This showed some effectiveness in the enhancement of a tumour-specific immune response but failed to eradicate the tumour.
- TbR transforming growth factor beta receptor
- Galunisertib which has been tested as a monotherapy or in combination with alkylating agents, Lomustine or temozolamide for glioblastoma and other combinations. These approaches have focussed on the inhibitory microenvironment and have not been particularly effective.
- the present inventors have designed and generated cells with an in-built mechanism to reduce immunosupprossive signalling by molecules such as TQRb.
- the present invention provides engineered cells comprising a dominant negative SMAD which renders the cells less susceptible (i.e. more resistant) to TGFb-mediated signalling.
- the present invention provides a cell which co-expresses (i) a dominant-negative SMAD (dnSMAD); and (ii).a chimeric antigen receptor (CAR) or a transgenic T-cell receptor (TCR).
- dnSMAD dominant-negative SMAD
- CAR chimeric antigen receptor
- TCR transgenic T-cell receptor
- the dnSMAD may be a dominant negative SMAD2, SMAD3 or SMAD4 polypeptide.
- the dnSMAD may lack a functional MH1 domain and/or a functional nuclear localization signal (NLS) domain.
- the present invention provides a chimeric dominant negative SMAD (dnSMAD) which comprises at least two dnSMAD polypeptides.
- dnSMAD chimeric dominant negative SMAD
- the at least two dnSMAD polypeptides may be joined by a linker domain.
- the chimeric dnSMAD may comprise: (i) a dnSMAD2 polypeptide and a dnSMAD3 polypeptide; (ii) a dnSMAD2 polypeptide and a dnSMAD4 polypeptide; or (iii) a dnSMAD3 polypeptide and a dnSMAD4 polypeptide.
- a polynucleotide which encodes a chimeric dnSMAD according to the second aspect of the invention.
- a nucleic acid construct which comprises: (i) a first polynucleotide according to the third aspect of the invention or which encodes a dnSMAD as defined in the first aspect of the invention; and (ii) a second polynucleotide which encodes a chimeric antigen receptor (CAR) or a transgenic T-cell receptor (TCR).
- CAR chimeric antigen receptor
- TCR transgenic T-cell receptor
- the first and second polynucleotides may be separated by a co-expression site.
- the present invention provides a kit of polynucleotides comprising: (i) a first polynucleotide according to the third aspect of the invention or which encodes a dnSMAD as defined in the first aspect of the invention; and (ii) a second polynucleotide which encodes a chimeric antigen receptor (CAR) or a transgenic T-cell receptor (TCR).
- a kit of polynucleotides comprising: (i) a first polynucleotide according to the third aspect of the invention or which encodes a dnSMAD as defined in the first aspect of the invention; and (ii) a second polynucleotide which encodes a chimeric antigen receptor (CAR) or a transgenic T-cell receptor (TCR).
- CAR chimeric antigen receptor
- TCR transgenic T-cell receptor
- the present invention provides a vector which comprises a polynucleotide according to the second aspect of the invention or a nucleic acid construct according to the third aspect of the invention.
- kits of vectors which comprises: (i) a first vector which comprises a polynucleotide which encodes a dnSMAD as defined above or a chimeric dnSMAD according to the second aspect of the invention; and (ii) a second vector which comprises a polynucleotide which encodes a chimeric antigen receptor (CAR) or a transgenic T-cell receptor (TCR).
- CAR chimeric antigen receptor
- TCR transgenic T-cell receptor
- the present invention provides a pharmaceutical composition which comprises a plurality of cells according to the first aspect of the invention.
- the present invention provides a pharmaceutical composition according to the eighth aspect of the invention, for use in treating a disease.
- the present invention provides a method for treating a disease, which comprises the step of administering a pharmaceutical composition according to the eighth aspect of the invention to a subject in need thereof.
- the present invention provides the use of a cell according to the first aspect of the invention in the manufacture of a pharmaceutical composition for the treatment of a disease.
- the disease may be cancer.
- a method for making a cell according to the first aspect of the invention which comprises the step of introducing: a polynucleotide according to the third aspect of the invention, a nucleic acid construct according to the fourth aspect of the invention, a kit of polynucleotides according to the fifth aspect of the invention, a vector according to the sixth aspect of the invention, or a kit of vectors according to the seventh aspect of the invention into a cell ex vivo.
- the present inventors provide engineered immune effector cells which counteract the inhibitory microenvironment and prevent inhibition of immune effector cells, thereby augmenting the ability of intrinsic tumour-specific T cells or engineered immune effector cells to attack the tumour.
- FIG. 1 Schematic diagram showing different generations of chimeric antigen receptors. The basic architecture of a canonical CAR is shown as well as different iterations of the three generations of this form of receptor.
- FIG. 2 Schematic diagram showing the domain structure of SMAD2, SMAD3, SMAD4 and SMAD7.
- FIG. 3 Schematic diagram showing a dnSMAD2/3 preventing signalling through the TQRb receptor.
- FIG. 4 Schematic diagram showing a dnSMAD4 preventing signalling through the TQRb receptor.
- FIG. 5 Cytotoxicity assay.
- T cells expressing a second generation anti-GD2 CAR with a CD28-CD3z endodomain either alone or in combination with a truncated SMAD2 (dnSMAD2-MH2) or truncated SMAD3 (dnSMAD3-MH2) molecule were co-cultured with GD2-expressing SupT1 target cells at a 2:1 and 4:1 target: effector cell ratio.
- TQRb was spiked into the co-culture at 10ng/ml on day 0 and target cell killing was investigated at day 5 and 7 by flow cytometry.
- TGF-b Transforming growth factor beta
- the transforming growth factor beta receptors are a superfamily of serine/threonine kinase receptors. These receptors bind members of the T ⁇ Rb superfamily of growth factor and cytokine signalling proteins. There are five type II receptors (which are activatory receptors) and seven type I receptors (which are signalling propagating receptors). Type I receptors are also known as activin receptor-like kinases (ALKS).
- Auxiliary co-receptors also known as type III receptors
- T ⁇ Rb superfamily of ligands binds to type I and type II receptors.
- the amino acid sequence of human TGF-beta receptor type-1 O ⁇ RI is available from UniProt accession P36897.
- the amino acid sequence of human TGF-beta receptor type-2 (TbRII) is available from UniProt accession P37173.
- T ⁇ Rb1 and 2 are implicated in cancer, where they may stimulate the cancer stem cell, increase fibrosis / desmoplastic reactions and suppress immune recognition of the tumour.
- T ⁇ Rb cancerous tumour cells
- T ⁇ Rb can be produced by the wide variety of non- cancerous cells present at the tumour site.
- tumour-associated T cells natural killer (NK) cells
- macrophages macrophages
- epithelial cells stromal cells
- T ⁇ Rb1 , 2 and 3 signal via binding to receptors TbRII and then association to TbRI and in the case of T ⁇ Rb2 also to TbRIII. This leads to subsequent signalling through SMADs via TbRI.
- TGFBRI and TGFBRII are composed of a short disulphide rich endodomain that is heavily bonded in a three-finger toxin conformation. These domains contain binding sites for serine/threonine kinases.
- TGF-b ligation to TGFBRII the endodomain is auto-phosphorylated enabling the recruitment and subsequent cross-linking with TGFBRI.
- TGF-b receptors are serine/threonine kinases. Following TGF-b ligation, TGFBRII is auto- phosphorylated enabling the recruitment and cross-linking with TGFBRI. Activation of TGFBRI kinases phosphorylates receptor regulated SMAD (R-SMAD) proteins.
- R-SMAD receptor regulated SMAD
- R-SMADs are transcription factors specific for the TGF-b family. Upon receptor ligation, serine residues in the C-terminus of R-SMAD are phosphorylated resulting in their detachment from the TGFBRI- TGFBRII receptor complex. R-SMADs exist in two subtypes with SMAD 2 and SMAD 3 phosphorylated upon TGF-b receptor ligation. Once phosphorylated, both R-SMAD subclasses form a complex with SMAD 4 which is a common mediator SMAD (co-SMAD).
- SMAD proteins consist of two globular domains connected by a linker region.
- the C-terminal domain, or MFI2 domain mediates protein-protein interaction with numerous regulator and effector proteins, including the TbB receptors, certain cytoplasmic anchor proteins, lineage-specific DNA-binding cofactors, and chromatin modifiers.
- T ⁇ Bb R-SMAD is phosphorylated by T ⁇ Bb receptor. This phosphorylation targets two serine residues in the SMAD C terminus sequence, pSer-X-pSer, creating an acidic tail that drives the formation of SMAD transcriptional complexes.
- Phosphorylated R-SMAD proteins bind the MH2 domain of SMAD 4 enabling accumulation of this complex at the nucleus, where SMAD complexes act as DNA site-specific transcriptional regulators.
- SMAD 3 and SMAD 4 recognise 5’CAGAC3’ binding elements.
- the regulation of gene expression is achieved by SMAD proteins interacting with transcriptional activators and repressors to initiate the functions associated with TGF-b ligands.
- dominant-negative SMAD (dnSMAD) polypeptide refers to a SMAD which acts antagonistically to the wild-type SMAD polypeptide.
- a dnSMAD polypeptide may be capable of binding to TQBbR and/or to other SMADs but be incapable of modulating or have a reduced capacity to modulate (e.g. inhibit/augment) transcriptional activity.
- a dnSMAD polypeptide may have an MH2 domain which is capable of binding to TQBbR and to other SMADs but is incapable of modulating or have a reduced capacity to modulate (e.g. inhibit/augment) transcriptional activity.
- a dnSMAD or chimeric dnSMAD as disclosed herein may inhibit the function of its natural or wild-type counterpart.
- the dnSMAD or chimeric dnSMADs may inhibit signalling induced by wild-type TQRb and thus neutralise its biological effects.
- Binding of the dnSMAD to Tb ⁇ or to other SMADs may inhibit signalling downstream of Tb ⁇ T
- Any method known in the art for determining protei protein interactions may be used to determine whether a dnSMAD is capable of binding to a TQBbR or to other SMAD(s). For example, co-immunoprecipitation followed by western blot.
- a SMAD is capable of modulating (e.g. initiating or augmenting) transcriptional activity.
- a transcriptional response assay may be used wherein cells are transfected with a promoter reporter construct e.g. luciferase in the presence of various combinations of SMAD plasmids. The activity of the reporter gene may be measured as an indicator of SMAD ability to initiate transcriptional activity.
- a dnSMAD may be identified or characterised using a TQBb cytotoxicity assay.
- TQBb cytotoxicity assay An example of methodology for such an assay is as follows: PBMCs depleted of CD56+ cells and transfected with nucleic acids encoding dnSMADs or wild-type SMADs may be co-cultured with target cells in e.g. a 1 :2 effector: target cell ratio in the presence and absence of TQBb. Cytotoxicity may be assessed by flow cytometry e.g. after 1 , 5 and 7 days. The dnSMAD may show increased (e.g.
- the dnSMAD reduces (e.g. inhibits) the effects of TQBb.
- a dnSMAD may be identified or characterised using a proliferation assay.
- An example of methodology for such an assay is as follows: T cells transfected with nucleic acids encoding dnSMADs or wild-type SMADs may be co-cultured with target cells in e.g. a 1 :2 effector: target cell ratio in the presence and absence of TQRb.
- the dnSMAD may restore proliferation compared with a wild-type SMAD in the presence of TQRb.
- the proliferation rate of the cell comprising the dnSMAD in the presence of TQRb may be essentially the same as the proliferation rate of a cell comprising the dnSMAD in the absence of TQRb e.g.
- the difference in proliferation rate between a cell comprising a dnSMAD in the presence of TQRb compared with proliferation rate of a cell comprising a dnSMAD the absence of TQRb may be less than 40% difference, less than 30% difference, less than 20% difference, less than 20% difference, less than 10% difference, less than 5% difference or less than 3% difference.
- the dnSMAD may be a dominant negative version of SMAD2, SMAD3 or SMAD4.
- the dnSMAD may be a dominant negative SMAD2 polypeptide.
- the dnSMAD may be a dominant negative SMAD3 polypeptide.
- the dnSMAD may be a dominant negative SMAD4 polypeptide.
- the dnSMAD may lack a functional MH1 domain.
- An“MH1 domain” as used herein refers to a conserved MAD homology domain of at the N terminus of a SMAD protein.
- the MH1 domain is capable of DNA binding.
- the MH1 domain negatively regulates the functions of the MH2 domain.
- DNA binding may be determined using any method known in the art, for example quantitative-electrophoretic mobility shift assay (EMSA) with labelled DNA and purified protein e.g. purified MH1 protein.
- ESA quantitative-electrophoretic mobility shift assay
- Other methods for measuring DNA binding include isothermal titration calorimetry which titrates small volumes of concentrated ligand (e.g. MH1 ) into the DNA and measures the amount of heat which needs to be added or subtracted to return it to a reference temperature.
- the MH1 domain of SMAD2 corresponds to amino acids 10-176 of SEQ ID NO: 1.
- SEQ ID NO:1 which is UniProtKB accession Q15796 (SMAD2 Human).
- RQTVTSTPCWIELHLNGPLQWLDKVLTQMGSPSVRCSSMS (SEQ ID NO: 1 ).
- SEQ ID NO: 2 is UniProtKB accession P84022 (SMAD3_Human).
- SMAD 4 corresponds to amino acids 18-142 of SEQ ID NO: 3.
- SEQ ID NO: 3 is UniProtKB accession Q13485 (SMAD4_Human). MDNMSITNTPTSNDACLSIVHSLMCHRQGGESETFAKRAIESLVKKLKEKKDELDSLITAITT
- IADPQPLD SEQ ID NO: 3
- the phrase“corresponds to” indicates that the amino acid position is equivalent to the one shown for the recited amino acid sequence e.g. the amino acid sequence shown in SEQ ID NO: 1 . It will be appreciated that the actual number of amino acids from the N-terminus of the protein may vary between different proteins. However, it is clear from, an alignment of the dnSMAD protein or chimeric dnSMAD protein with the sequence of the recited amino acid sequence (e.g. SEQ ID NO: 1 ) which is the“equivalent” amino acid position.
- S-X-S motif at amino acid positions 465-467 of SEQ ID NO: 1 correspond to amino acid positions ! 92-194 of SEQ ID NO: 4 and SEQ ID NO: 8.
- a functional MH1 domain is capable of DNA binding.
- a polypeptide which“lacks a functional MH1 domain” as used herein exhibits reduced DNA binding or DNA binding is completely eliminated when compared with a wild type MH1 domain.
- a polypeptide which lacks a functional MH1 domain may exhibit less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10% of the DNA binding compared to a wild type MH1 polypeptide.
- a polypeptide which lacks a functional MH1 domain may be unable to bind to DNA.
- the dnSMAD essentially lacks a MH1 domain compared to a corresponding wild-type SMAD polypeptide.
- the nucleic acid sequence which encodes the MH1 domain may be wholly or partially deleted.
- the deletion may be continuous, or may comprise a plurality of sections of sequence.
- the deletion preferably removes a sufficient amount of nucleotide sequence such that the nucleic acid sequence no longer encodes a functional protein.
- the deletion may be total, in which case 100% of the coding portion of the nucleic acid sequence is absent, when compared to the corresponding nucleic acid sequence of a wild type MH1 .
- the deletion may, for example, remove at least 70%, 80%, 90% or 95% of the MH1 -coding portion of the nucleic acid sequence.
- the dnSMAD may lack a functional nuclear localization signal (NLS) domain.
- nuclear localisation signal has its normal meaning in the art and refers to an amino acid sequence which tags a protein for nuclear transport for import into the cell nucleus.
- NLS typically comprise one or more short sequences of charged lysines or arginines.
- a polypeptide which lacks a functional NLS is not capable of import to the cell nucleus. Any methods known in the art for monitoring translocation of proteins may be used to determine whether a polypeptide lacks a functional NLS. For example, nuclear import may be monitored using immunofluorescence confocal microscopy or by subcellular fractionation and organelle isolation of the nucleus followed by western blot to determine the subcellular localisation of the polypeptide.
- the wild-type SMAD NLS may have the sequence: Lys-Lys-Leu-Lys-Lys.
- the NLS sequence in SMAD3 spans residues 40-44 and has the sequence: KKLKK
- the dnSMAD lacks a NLS domain compared to a corresponding wild- type SMAD polypeptide.
- the dnSMAD may comprise a deletion or substitution in the NLS.
- one or more of the lysine residues in the NLS may be deleted or substituted with another amino acid such as glutamine. All four lysine residues may be substituted with an alternative amino acid.
- MFI2 domain refers to a conserved MAD homology domain of at the C terminus of a SMAD protein.
- the MFI2 domain contain a central b-sandwich with a conserved loop-helix is capable of binding phosphor-serine residues.
- the MFI2 domain mediates protein: protein interactions with regulator and effector proteins, including the Tb ⁇ receptors, cytoplasmic anchor proteins, lineage-specific DNA-binding cofactor and chromatin modifiers.
- the dnSMAD may comprise, consist essentially of, or consist of a wild-type MH2 domain.
- the dnSMAD may consist essentially of or consist of the MH2 domain of wild- type SMAD2 which has the sequence shown as SEQ ID No. 4.
- the dnSMAD may consist essentially of or consist of the MH2 domain of wild- type SMAD3 which has the sequence shown as SEQ ID No. 8.
- the dnSMAD may consist essentially of or consist of the MH2 domain of wild- type SMAD4 which has the sequence shown as SEQ ID No. 12.
- the dnSMAD may comprise, consist essentially of or consist of a truncated MH2 domain of a wild type SMAD polypeptide, such as a truncated version of SEQ ID NO: 4, 8 or 12.
- the truncation may be at the C-terminus, for example, Yuan and Varga (2001 , as above) describe a dnSMAD4 with a C-terminal truncation of 51 amino acids.
- the truncated MH2 domain may:
- the truncated MH2 domain may:
- a dnSMAD which lacks a functional MH1 domain may further:
- a) lack at least the amino acids which correspond to position 457 to 467 of SEQ ID NO: 1 ; or b) lack at least the amino acids which correspond to position 444 to 467 of SEQ ID NO: 1.
- a dnSMAD4 may comprise the mutation R497H, K507Q and/or R515G where the amino acid numbering corresponds to the numbering set forth in SEQ ID NO: 3.
- a dnSMAD3 may comprise a mutation K378R and/or K314R where the amino acid numbering corresponds to the numbering set forth in SEQ ID NO: 2.
- the truncation may be a deletion of at least 1 1 amino acids from the C terminus, wherein the amino acid numbering corresponds to SEQ ID NO: 4.
- the dnSMAD may consist essentially of or consist of SEQ ID NO: 5 or a variant of SEQ ID NO: 5 which has at least 80% (preferably at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%) sequence identity thereto.
- the truncation may be a deletion of at least 24 amino acids from the C terminus, wherein the amino acid numbering corresponds to SEQ ID NO: 4.
- the dnSMAD may consist essentially of or consist of SEQ ID NO: 6 or a variant of SEQ ID NO: 6 which has at least 80% (preferably at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%) sequence identity thereto.
- the truncation may be a deletion of at least 1 1 amino acids from the C terminus, wherein the amino acid numbering corresponds to SEQ ID NO: 8.
- the dnSMAD may consist essentially of or consist of SEQ ID NO: 9 or a variant of SEQ ID NO: 9 which has at least 80% (preferably at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%) sequence identity thereto.
- the truncation may be a deletion of at least 24 amino acids from the C terminus, wherein the amino acid numbering corresponds to SEQ ID NO: 8.
- the dnSMAD may consist essentially of or consist of SEQ ID NO: 10 or a variant of SEQ ID NO: 10 which has at least 80% (preferably at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%) sequence identity thereto.
- the dnSMAD may further comprise a mutation in the MH2 domain which increases the binding affinity of the dnSMAD for a phosphorylated TQEb receptor compared to a corresponding MH2 domain which does not comprise said mutation.
- An increase in binding to the phosphorylated TQEb may be determined using any method known in the art or described herein; for example cells may be transfected with a nucleic acid encoding a dnSMAD comprising a mutation in the MH2 domain or with a nucleic acid encoding a corresponding MH2 domain which does not comprise said mutation.
- a comparison of the binding to phosphorylated TQRb may be determined by co- immunoprecipitation and western blotting of the phosphorylated TQRb receptor and the dnSMAD versus co-immunoprecipitation of the phosphorylated TQRb receptor an MH2 domain which does not comprise the mutation.
- radioactive ligand binding assays including saturation binding, scatchard plot
- non-radioactive ligand binding assays including fluorescence polarization, fluorescence resonance energy transfer and surface plasmon resonance/Biacore
- solid phase ligand binding assays include fluorescence polarization, fluorescence resonance energy transfer and surface plasmon resonance/Biacore, and solid phase ligand binding assays.
- the dnSMAD may comprises a substitution of one or both amino acids which correspond to positions 465 and 467 of SEQ ID NO: 1 .
- the substitution of the amino acids which correspond to positions 465 and 467 of SEQ ID NO: 1 may prevent phosphorylation of the SMAD C terminus sequence.
- the substitution of the amino acids which correspond to positions 465 and 467 of SEQ ID NO: 1 may replace one or both of the serine residues in the Ser-X-Ser motif with an amino acid which cannot be phosphorylated.
- one or both serine residues are not substituted for threonine, tyrosine, histidine, lysine, arginine, aspartic acid or glutamic acid.
- one or both serine residues are substituted for alanine.
- the dnSMAD may consist essentially of or consist of SEQ ID NO: 7 or a variant of SEQ ID NO: 7 which has at least 80% (preferably at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%) sequence identity thereto and comprises amino acid substitutions at positions which correspond to positions 465 and 467 of SEQ ID NO: 1.
- One or both of the substitutions may be the substitution of serine for an amino acid which cannot be phosphorylated (e.g. alanine).
- the dnSMAD may consist essentially of or consist of SEQ ID NO: 1 1 or a variant of SEQ ID NO: 1 1 which has at least 80% (preferably at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%) sequence identity thereto and comprises amino acid substitutions at positions which correspond to positions 465 and 467 of SEQ ID NO: 1.
- one or both of the substitutions may be the substitution of serine for an amino acid which cannot be phosphorylated (e.g. alanine).
- the present invention provides a chimeric dnSMAD which comprises at least two dnSMAD polypeptides as defined herein.
- the chimeic dnSMAD may comprise at least two, at least 3 at least 4, at least five dnSMADs as defined herein.
- the chimeric dnSMAD comprises a dnSMAD2 and a dnSMAD3. In one embodiment the chimeric dnSMAD comprises a dnSMAD2 and a dnSMAD4. In one embodiment the chimeric dnSMAD comprises a dnSMAD3 and a dnSMAD4. In one embodiment the chimeric dnSMAD comprises at least two dnSMAD2s. In one embodiment the chimeric dnSMAD comprises at least two dnSMAD2s. In one embodiment the chimeric dnSMAD comprises at least two dnSMAD4s.
- the chimeric dnSMAD comprises a dnSMAD2, a dnSMAD3 and a dnSMAD4.
- the dnSMAD polypeptides of the chimeric dnSMAD may be connected by a linker domain.
- Linkers are short sequences which separate multiple domains in a protein.
- a linker may comprise flexible residues such as glycine and serine repeats which allow the adjacent protein domains to move relative to one another.
- the chimeric dnSMAD may comprise one or more serine-glycine linker domains.
- the linker may be of any length with provides sufficient flexibility.
- the linker may be 2, 3, 4, 5, 10, 15, 20, 25, 30 or more residues long.
- the chimeric dnSMAD comprises a linker comprising or consisting essentially of or consisting of the following sequence: LEYSGGGSGGGSLE (SEQ ID NO: 19).
- the chimeric dnSMAD comprises a dnSMAD2 polypeptide and a dnSMAD3 polypeptide.
- the chimeric dnSMAD may comprise an SMAD2-MH2 domain and a SMAD3-MH2 domain.
- one or more of the SMAD-MH2 domains may be a truncated MH2 domain.
- the dnSMAD may also comprise a linker domain.
- the dnSMAD chimeric molecule comprises or consists essentially of or consists of a sequence set forth in SEQ ID NO: 13.
- the linker sequence is shown in bold.
- the chimeric dnSMAD may comprise a dnSMAD2 polypeptide and a dnSMAD4 polypeptide.
- one or more of the SMAD-MH2 domains may be a truncated MH2 domain.
- the dnSMAD may also comprise a linker domain.
- the dnSMAD chimeric molecule comprises or consists essentially of or consists of a sequence set forth in SEQ ID NO: 14.
- the linker sequence is shown in bold.
- the chimeric dnSMAD may comprise a dnSMAD3 polypeptide and a dnSMAD4 polypeptide.
- one or more of the SMAD-MH2 domains may be a truncated MH2 domain.
- the dnSMAD may also comprise a linker domain.
- the dnSMAD chimeric molecule comprises or consists essentially of or consists of a sequence set forth in SEQ ID NO: 15.
- the linker sequence is shown in bold.
- the chimeric dnSMAD may comprise a dnSMAD2 polypeptide and a dnSMAD3 polypeptide.
- one or more of the SMAD-MH2 domains may comprise a mutation in the SXS motif of the MH2 domain.
- the dnSMAD may also comprise a linker domain.
- the dnSMAD chimeric molecule comprises or consists essentially of or consists of a sequence set forth in SEQ ID NO: 16.
- the linker sequence is shown in bold.
- the chimeric dnSMAD may comprise a dnSMAD2 polypeptide and a dnSMAD4 polypeptide.
- the SMAD2-MH2 domain may comprise a mutation in the SXS motif of the MH2 domain.
- the dnSMAD may also comprise a linker domain.
- the dnSMAD chimeric molecule comprises or consists essentially of or consists of a sequence set forth in SEQ ID NO: 17.
- the linker sequence is shown in bold.
- the chimeric dnSMAD may comprise a dnSMAD3 polypeptide and a dnSMAD4 polypeptide.
- the SMAD3-MH2 domain may comprise a mutation in the SXS motif of the MH2 domain.
- the dnSMAD may also comprise a linker domain.
- the dnSMAD chimeric molecule comprises or consists essentially of or consists of a sequence set forth in SEQ ID NO: 18.
- the linker sequence is shown in bold.
- a chimeric dnSMAD according to the present invention may comprise at least one dnSMAD according to the present invention.
- a dnSMAD may comprise at least one sequence selected from: SEQ ID NO: 4 to 18; or a variant having at least 80% (preferably at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%) sequence identity to SEQ ID NO: 4 to 18.
- the chimeric dnSMAD may comprise two, or three or four or five or more sequences selected from SEQ ID NO: 4 to 18; or a variant having at least 80% (preferably at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%) sequence identity to SEQ ID NO: 4 to 18.
- the present invention provides a cell which encodes a dnSMAD, wherein the expression of said dnSMAD may be“tunable”.
- “tunable” means that it is possible to increase, decrease, turn on or turn off the expression of the dnSMAD in the engineered immune effector cell.
- expression of the dnSMAD may be controlled or tuned by an inducible promoter.
- the dnSMAD may be regulated by Nuclear factor of activated T cells (NFAT) response element.
- An NFAT response element may comprise the nucleotide sequence set forth in SEQ ID NO: 20 or a variant thereof.
- Variant sequences of SEQ ID NO: 20 may have at least 80%, 85%, 90%, 95%, 98% or 99% sequence identity to SEQ ID NO: 20.
- the variant sequence is able to function as a NFAT response element.
- the NFAT response element may comprise repeat units such as 3, 4, 5 or 6 repeat units.
- the NFAT response element may comprise 3, 4, 5 or 6 repeat units of SEQ ID NO: 20.
- the NFAT response element may be positioned in front of a promoter (e.g. a CMV promoter).
- the expression or activity of the dnSMAD may be controlled or tuned through interaction with an intracellular retention domain.
- the dnSMAD may be retained within a cellular compartment by interaction with an intracellular retention domain.
- An agent may be used to disrupt the interaction with the intracellular retention domain, thereby allowing translocation of the dnSMAD and expression of the dnSMAD in the appropriate cellular localisation of the engineered immune effector cell.
- the activity of the dnSMAD factor may be controlled or tuned.
- the dnSMAD or chimeric dnSMAD according to the present invention may compete with the wild type SMAD protein for the receptor-docking domain on TbR or for competes with the wild type SMAD protein for binding with partner proteins.
- the dnSMAD or chimeric dnSMAD according to the present invention may reduce or completely inhibit TQRb signalling in the engineered immune effector cell.
- the dnSMAD or chimeric snSMAD according to the present invention may reduce or eliminate signalling downstream of Tb ⁇ T
- Assays for measuring downstream signalling of Tb ⁇ are known in the art such as luminescent kinase assays which measure ADP formed from the kinase reaction or measuring the proportion of cytoplasmic signalling molecules such as SMAD/SMAD2 phosphorylation. Any method known in the art may be used to measure downstream signalling of Tb ⁇ T dnSMADs or chimeric SMADs according to the present invention may maintain their ability to bind to TbR and/or to other SMADs. The ability of a variant TQRb to bind to a TbR or to another SMAD may be measured by any means known in the art for example by an ELISA assay, to detect TbR receptor chain or other SMADs.
- dnSMADs may block the binding of wild-type SMADs to TbR or to other wild type SMADs. This may be measured by a competitive ELISA for example, by covering the plates with TbR or wild type SMAD and assessing the ability of the dnSMAD (or chimeric dnSMAD) to inhibit binding of wild type TbR or wild type SMADs to their wild type binders. dnSMADs may be tested for in vitro binding to consensus binding sequences for example using GST fusion proteins.
- dnSMADs may have reduced ability to signal through TbR or may not be capable of signalling through Tb ⁇ T This may be measure by measuring the transcription of genes induced by TQRb for example by qPCR.
- a dnSMAD (or chimeric dnSMADs) according to the present invention may exhibit reduced phosphorylation of the wild type SMAD2/3. This may be measured by western blot or flow- cytometry using antibodies specific for the phosphorylated tyrosine of the wild-type SMAD.
- dnSMADs (or chimeric dnSMADs) according to the present invention may be capable of inhibiting signalling induced by wild-type TQRb. This may be measured by Western immunoblotting assays and quantifying the levels of phosphorylated SMAD2 and SMAD3 in cell lysates treated with the mutant or wild-type TQRb.
- the present invention relates to a cell which expresses a dominant-negative SMAD (dnSMAD).
- dnSMAD dominant-negative SMAD
- the cell may also express.
- CAR chimeric antigen receptor
- TCR transgenic T- cell receptor
- the cell may be engineered to express the dnSMAD and/or CAR/TCR.
- An“engineered cell” as used herein means a cell which has been modified to comprise or express a nucleic acid sequence which is not naturally encoded by the cell.
- Methods for engineering cells include but are not limited to genetic modification of cells e.g. by transduction such as retroviral or lentiviral transduction, transfection (such as transient transfection - DNA or RNA based) including lipofection, polyethylene glycol, calcium phosphate and electroporation. Any suitable method may be used to introduce a nucleic acid sequence into a cell.
- nucleic acid sequence encoding the dnSMAD is not naturally expressed by a corresponding, unmodified cell.
- An engineered cell is a cell whose genome has been modified e.g. by transduction or by transfection, such as retroviral or lentiviral transduction.
- the term“introduced” refers to methods for inserting foreign DNA or RNA into a cell.
- the term introduced includes both transduction and transfection methods.
- Transfection is the process of introducing nucleic acids into a cell by non-viral methods.
- Transduction is the process of introducing foreign DNA or RNA into a cell via a viral vector.
- Engineered cells according to the present invention may be generated by introducing DNA or RNA coding a dnSMAD by one of many means including transduction with a viral vector, transfection with DNA or RNA.
- Cells may be activated and/or expanded prior to the introduction of a nucleic acid sequence encoding the dnSMAD, for example by treatment with an anti-CD3 monoclonal antibody or both anti-CD3 and anti-CD28 monoclonal antibodies.
- activated means that a cell has been stimulated, causing the cell to proliferate, differentiate or initiate an effector function.
- Methods for measuring cell activation include, for example, measuring the expression of activation markers by flow cytometry, such as the expression of CD69, CD25, CD38 or HLA-DR or measuring intracellular cytokines.
- expansion means that a cell or population of cells has been induced to proliferate.
- the expansion of a population of cells may be measured for example by counting the number of cells present in a population.
- the phenotype of the cells may be determined by methods known in the art such as flow cytometry.
- An“immune effector cell” as used herein is a cell of the immune system which responds to a stimulus and effects a change.
- an immune effector cell may a T cell (such as an alpha-beta T cell or a gamma- delta T cell), a B cell (such as a plasma cell), a Natural Killer (NK) cell or a macrophage.
- T cell such as an alpha-beta T cell or a gamma- delta T cell
- B cell such as a plasma cell
- NK Natural Killer
- Cytolytic immune cell as used herein is a cell which directly kills other cells. Cytolytic cells may kill cancerous cells; virally infected cells or other damaged cells. Cytolytic immune cells include T cells and Natural killer (NK) cells.
- Cytolytic immune cells can be T cells or T lymphocytes which are a type of lymphocyte that play a central role in cell-mediated immunity.
- T cells can be distinguished from other lymphocytes, such as B cells and NK cells, by the presence of a TCR on their cell surface.
- Cytolytic T cells destroy virally infected cells and tumour cells, and are also implicated in transplant rejection.
- CTLs express the CD8 at their surface.
- CTLs may be known as CD8+ T cells. These cells recognize their targets by binding to antigen associated with MHC class I, which is present on the surface of all nucleated cells.
- MHC class I MHC class I
- IL-10 adenosine and other molecules secreted by regulatory T cells, the CD8+ cells can be inactivated to an anergic state, which prevent autoimmune diseases such as experimental autoimmune encephalomyelitis.
- the cell of the present invention may be a T -cell, such as an alpha-beta T cell or a gamma- delta T cell.
- Natural Killer Cells are a type of cytolytic cell which form part of the innate immune system. NK cells provide rapid responses to innate signals from virally infected cells in an MHC independent manner.
- NK cells (belonging to the group of innate lymphoid cells) are defined as large granular lymphocytes (LGL) and constitute the third kind of cells differentiated from the common lymphoid progenitor generating B and T lymphocytes. NK cells are known to differentiate and mature in the bone marrow, lymph node, spleen, tonsils and thymus where they then enter into the circulation.
- LGL large granular lymphocytes
- the cell of the present invention may be a wild-type killer (NK) cell or a cytokine induced killer cell.
- the cell may be derived from a patient’s own peripheral blood (1 st party), or in the setting of a haematopoietic stem cell transplant from donor peripheral blood (2nd party), or peripheral blood from an unconnected donor (3rd party).
- T or NK cells for example, may be activated and/or expanded prior to being transduced with nucleic acid molecule(s) encoding the polypeptides of the invention, for example by treatment with an anti-CD3 monoclonal antibody.
- the cell may be derived from ex vivo differentiation of inducible progenitor cells or embryonic progenitor cells to T cells.
- an immortalized T-cell line which retains its lytic function may be used.
- the cell may be a haematopoietic stem cell (HSC).
- HSCs can be obtained for transplant from the bone marrow of a suitably matched donor, by leukapheresis of peripheral blood after mobilization by administration of pharmacological doses of cytokines such as G-CSF [peripheral blood stem cells (PBSCs)], or from the umbilical cord blood (UCB) collected from the placenta after delivery.
- cytokines such as G-CSF [peripheral blood stem cells (PBSCs)]
- PBSCs peripheral blood stem cells
- URB umbilical cord blood
- the marrow, PBSCs, or UCB may be transplanted without processing, or the HSCs may be enriched by immune selection with a monoclonal antibody to the CD34 surface antigen.
- Classical CARs are chimeric type I trans-membrane proteins which connect an extracellular antigen-recognizing domain (binder) to an intracellular signalling domain (endodomain).
- the binder is typically a single-chain variable fragment (scFv) derived from a monoclonal antibody (mAb), but it can be based on other formats which comprise an antibody-like antigen binding site or on a ligand for the target antigen.
- mAb monoclonal antibody
- a spacer domain may be necessary to isolate the binder from the membrane and to allow it a suitable orientation.
- a common spacer domain used is the Fc of lgG1. More compact spacers can suffice e.g. the stalk from CD8a and even just the lgG1 hinge alone, depending on the antigen.
- a trans membrane domain anchors the protein in the cell membrane and connects the spacer to the endodomain.
- TNF receptor family endodomains such as the closely related 0X40 and 4-1 BB which transmit survival signals.
- CARs have now been described which have endodomains capable of transmitting activation, proliferation and survival signals.
- CAR-encoding nucleic acids may be transferred to T cells using, for example, retroviral vectors.
- retroviral vectors In this way, a large number of antigen-specific T cells can be generated for adoptive cell transfer.
- the CAR binds the target-antigen, this results in the transmission of an activating signal to the T-cell it is expressed on.
- the CAR directs the specificity and cytotoxicity of the T cell towards cells expressing the targeted antigen.
- the antigen-binding domain is the portion of a classical CAR which recognizes antigen.
- Numerous antigen-binding domains are known in the art, including those based on the antigen binding site of an antibody, antibody mimetics, and T-cell receptors.
- the antigen-binding domain may comprise: a single-chain variable fragment (scFv) derived from a monoclonal antibody; a wild-type ligand of the target antigen; a peptide with sufficient affinity for the target; a single domain binder such as a camelid; an artificial binder single as a Darpin; or a single-chain derived from a T-cell receptor.
- scFv single-chain variable fragment
- tumour associated antigens are known, as shown in the following Table 3.
- the antigen-binding domain used in the present invention may be a domain which is capable of binding a TAA as indicated therein.
- the transmembrane domain is the sequence of a classical CAR that spans the membrane. It may comprise a hydrophobic alpha helix. The transmembrane domain may be derived from CD28, which gives good receptor stability.
- the CAR or transgenic TCR for use in the present invention may comprise a signal peptide so that when it is expressed in a cell, such as a T-cell, the nascent protein is directed to the endoplasmic reticulum and subsequently to the cell surface, where it is expressed.
- the core of the signal peptide may contain a long stretch of hydrophobic amino acids that has a tendency to form a single alpha-helix.
- the signal peptide may begin with a short positively charged stretch of amino acids, which helps to enforce proper topology of the polypeptide during translocation.
- At the end of the signal peptide there is typically a stretch of amino acids that is recognized and cleaved by signal peptidase.
- Signal peptidase may cleave either during or after completion of translocation to generate a free signal peptide and a mature protein. The free signal peptides are then digested by specific proteases.
- the receptor may comprise a spacer sequence to connect the antigen-binding domain with the transmembrane domain.
- a flexible spacer allows the antigen-binding domain to orient in different directions to facilitate binding.
- the spacer sequence may, for example, comprise an lgG1 Fc region, an lgG1 hinge or a human CD8 stalk or the mouse CD8 stalk.
- the spacer may alternatively comprise an alternative linker sequence which has similar length and/or domain spacing properties as an lgG1 Fc region, an lgG1 hinge or a CD8 stalk.
- a human lgG1 spacer may be altered to remove Fc binding motifs.
- the intracellular signalling domain is the signal-transmission portion of a classical CAR.
- CD3-zeta endodomain which contains 3 ITAMs. This transmits an activation signal to the T cell after antigen is bound.
- CD3-zeta may not provide a fully competent activation signal and additional co stimulatory signalling may be needed.
- chimeric CD28 and 0X40 can be used with CD3-Zeta to transmit a proliferative / survival signal, or all three can be used together.
- the intracellular signalling domain may be or comprise a T cell signalling domain.
- the intracellular signalling domain may comprise one or more immunoreceptor tyrosine- based activation motifs (ITAMs).
- ITAM immunoreceptor tyrosine- based activation motifs
- An ITAM is a conserved sequence of four amino acids that is repeated twice in the cytoplasmic tails of certain cell surface proteins of the immune system.
- the motif contains a tyrosine separated from a leucine or isoleucine by any two other amino acids, giving the signature YxxL/l.
- Two of these signatures are typically separated by between 6 and 8 amino acids in the tail of the molecule (YxxL/lx (6.8) YxxL/l).
- ITAMs are important for signal transduction in immune cells.
- the CD3 and z-chains of the T cell receptor complex are found in the tails of important cell signalling molecules such as the CD3 and z-chains of the T cell receptor complex, the CD79 alpha and beta chains of the B cell receptor complex, and certain Fc receptors.
- the tyrosine residues within these motifs become phosphorylated following interaction of the receptor molecules with their ligands and form docking sites for other proteins involved in the signalling pathways of the cell.
- the intracellular signalling domain component may comprise, consist essentially of, or consist of the O ⁇ 3-z endodomain, which contains three ITAMs.
- the O ⁇ 3-z endodomain transmits an activation signal to the T cell after antigen is bound.
- the intracellular signalling domain may comprise additional co-stimulatory signalling.
- 4-1 BB also known as CD137
- CD28 and 0X40 can be used with O ⁇ 3-z to transmit a proliferative / survival signal.
- the CAR may have the general format: antigen-binding domain-TCR element.
- TCR element means a domain or portion thereof of a component of the TCR receptor complex.
- the TCR element may comprise (e.g. have) an extracellular domain and/or a transmembrane domain and/or an intracellular domain e.g. intracellular signalling domain of a TCR element.
- the TCR element may selected from TCR alpha chain, TCR beta chain, a CD3 epsilon chain, a CD3 gamma chain, a CD3 delta chain, CD3 epsilon chain.
- T-cell receptor is a molecule found on the surface of T cells which is responsible for recognizing fragments of antigen as peptides bound to major histocompatibility complex (MHC) molecules.
- MHC major histocompatibility complex
- the TCR is a heterodimer composed of two different protein chains.
- the TCR consists of an alpha (a) chain and a beta (b) chain (encoded by TRA and TRB, respectively)
- the TCR consists of gamma and delta (g/d) chains (encoded by TRG and TRD, respectively).
- antigens recognized by the TCR can include the entire array of potential intracellular proteins, which are processed and delivered to the cell surface as a peptide/MHC complex.
- heterologous TCR molecules it is possible to engineer cells to express heterologous (i.e. non-native) TCR molecules by artificially introducing the TRA and TRB genes; or TRG and TRD genes into the cell using a vector.
- the genes for engineered TCRs may be reintroduced into autologous T cells and transferred back into patients for T cell adoptive therapies.
- Such‘heterologous’ TCRs may also be referred to herein as‘transgenic TCRs’.
- the transgenic TCR for use in the present invention may recognise a tumour associated antigen (TAA) when fragments of the antigen are complexed with major histocompatibility complex (MHC) molecules on the surface of another cell.
- TAA tumour associated antigen
- MHC major histocompatibility complex
- the transgenic TCR for use in the present invention may recognise a TAA listed in Table 3.
- the present invention provides a nucleic acid construct which comprises:
- the present invention also provides a kit comprising nucleic acid sequences according to the present invention.
- the kit may comprise
- polynucleotide As used herein, the terms“polynucleotide”,“nucleotide”, and“nucleic acid” are intended to be synonymous with each other.
- the nucleic acid construct may comprise a plurality of nucleic acid sequences which encode a dnSMAD or a chimeric dnSMAD according to the present invention; and a CAR or transgenic TCR.
- the nucleic acid construct may comprise two, three, four or more nucleic acid sequences which encode different components of the invention.
- the plurality of nucleic acid sequences may be separated by co-expression sites as defined herein.
- polynucleotides and nucleic acids can encode the same polypeptide as a result of the degeneracy of the genetic code.
- skilled persons may, using routine techniques, make nucleotide substitutions that do not affect the polypeptide sequence encoded by the polynucleotides described herein to reflect the codon usage of any particular host organism in which the polypeptides are to be expressed.
- the polynucleotides of the present invention are codon optimised to enable expression in a mammalian cell, in particular an immune effector cell as described herein.
- Nucleic acids according to the invention may comprise DNA or RNA. They may be single- stranded or double-stranded. They may also be polynucleotides which include within them synthetic or modified nucleotides. A number of different types of modification to oligonucleotides are known in the art. These include methylphosphonate and phosphorothioate backbones, addition of acridine or polylysine chains at the 3' and/or 5' ends of the molecule. For the purposes of the use as described herein, it is to be understood that the polynucleotides may be modified by any method available in the art. Such modifications may be carried out in order to enhance the in vivo activity or life span of polynucleotides of interest.
- variant in relation to a nucleotide sequence or amino acid sequence includes any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) nucleic acid(s) from or to the sequence.
- a co-expression site is used herein to refer to a nucleic acid sequence enabling co expression of nucleic acid sequences encoding the dnSMAD and a CAR or transgenic TCR according to the present invention.
- co-expression site between the first nucleic acid sequence and the second nucleic acid sequence.
- the same co-expression site may be used.
- the co-expression site may be a cleavage site.
- the cleavage site may be any sequence which enables the two polypeptides to become separated.
- the cleavage site may be self cleaving, such that when the polypeptide is produced, it is immediately cleaved into individual peptides without the need for any external cleavage activity.
- cleavage is used herein for convenience, but the cleavage site may cause the peptides to separate into individual entities by a mechanism other than classical cleavage.
- FMDV Foot-and-Mouth disease virus
- various models have been proposed for to account for the “cleavage” activity: proteolysis by a host-cell proteinase, autoproteolysis or a translational effect (Donnelly et al (2001 ) J. Gen. Virol. 82:1027-1041 ).
- the exact mechanism of such “cleavage” is not important for the purposes of the present invention, as long as the cleavage site, when positioned between nucleic acid sequences which encode proteins, causes the proteins to be expressed as separate entities.
- the cleavage site may be a furin cleavage site.
- Furin is an enzyme which belongs to the subtilisin-like proprotein convertase family. The members of this family are proprotein convertases that process latent precursor proteins into their biologically active products.
- Furin is a calcium-dependent serine endoprotease that can efficiently cleave precursor proteins at their paired basic amino acid processing sites. Examples of furin substrates include proparathyroid hormone, transforming growth factor beta 1 precursor, proalbumin, pro-beta-secretase, membrane type-1 matrix metalloproteinase, beta subunit of pro-nerve growth factor and von Willebrand factor.
- Furin cleaves proteins just downstream of a basic amino acid target sequence (canonically, Arg-X-(Arg/Lys)-Arg') and is enriched in the Golgi apparatus.
- the cleavage site may be a Tobacco Etch Virus (TEV) cleavage site.
- TSV Tobacco Etch Virus
- TEV protease is a highly sequence-specific cysteine protease which is chymotrypsin-like proteases. It is very specific for its target cleavage site and is therefore frequently used for the controlled cleavage of fusion proteins both in vitro and in vivo.
- the consensus TEV cleavage site is ENLYFQ ⁇ S (where‘V denotes the cleaved peptide bond).
- Mammalian cells such as human cells, do not express TEV protease.
- the present nucleic acid construct comprises a TEV cleavage site and is expressed in a mammalian cell - exogenous TEV protease must also expressed in the mammalian cell.
- the cleavage site may encode a self-cleaving peptide.
- A‘self-cleaving peptide’ refers to a peptide which functions such that when the polypeptide comprising the proteins and the self cleaving peptide is produced, it is immediately“cleaved” or separated into distinct and discrete first and second polypeptides without the need for any external cleavage activity.
- the self-cleaving peptide may be a 2A self-cleaving peptide from an aphtho- or a cardiovirus.
- the primary 2A/2B cleavage of the aptho- and cardioviruses is mediated by 2A “cleaving” at its own C-terminus.
- apthoviruses such as foot-and-mouth disease viruses (FMDV) and equine rhinitis A virus
- the 2A region is a short section of about 18 amino acids, which, together with the N-terminal residue of protein 2B (a conserved proline residue) represents an autonomous element capable of mediating“cleavage” at its own C-terminus (Donelly et al (2001 ) as above).
- 2A-like sequences have been found in picornaviruses other than aptho- or cardioviruses, ‘picornavirus-like’ insect viruses, type C rotaviruses and repeated sequences within Trypanosoma spp and a bacterial sequence (Donnelly et al., 2001 ) as above.
- the co-expression sequence may be an internal ribosome entry sequence (IRES).
- the co expressing sequence may be an internal promoter.
- the present invention also provides a vector, or kit of vectors which comprises one or more nucleic acid sequence(s) or nucleic acid construct(s) of the invention.
- a vector may be used to introduce the nucleic acid sequence(s) or construct(s) into a host cell so that it expresses a dnSMAD as defined herein.
- the vector may comprise a plurality of nucleic acid sequences which encode different components as provided by the present invention.
- the vector may comprise two, three, four or more nucleic acid sequences which encode different components, such as the dnSMAD and a CAR or transgenic TCR.
- the plurality of nucleic acid sequences may be separated by co-expression sites as defined herein.
- the vector may, for example, be a plasmid or a viral vector, such as a retroviral vector or a lentiviral vector, or a transposon based vector or synthetic mRNA.
- the vector may be capable of transfecting or transducing a cell.
- the present invention also relates to a pharmaceutical composition
- a pharmaceutical composition comprising an engineered immune effector cell according to the present invention or a cell obtainable (e.g. obtained) by a method according to the present invention.
- the present invention also provides a pharmaceutical composition
- a pharmaceutical composition comprising a nucleic acid construct according to the present invention, a first and second polynucleotide as defined herein, or a vector according to the present invention or a first and second vector as defined herein.
- the invention relates to a pharmaceutical composition containing a cell according to the present invention.
- the pharmaceutical composition may additionally comprise a pharmaceutically acceptable carrier, diluent or excipient.
- the pharmaceutical composition may optionally comprise one or more further pharmaceutically active polypeptides and/or compounds.
- Such a formulation may, for example, be in a form suitable for intravenous infusion.
- the present invention provides a method for treating and/or preventing a disease which comprises the step of administering an engineered immune effector cell according to the invention, or obtainable (e.g. obtained) by a method according to the present invention, or a nucleic acid construct according to the present invention, or a first and second nucleic acid sequence as defined herein; a vector according to the present invention or a first and second vector as described herein (for example in a pharmaceutical composition as described above) to a subject.
- the present methods for treating and/or preventing a disease may comprise administering an engineered immune effector cell according to the present invention (for example in a pharmaceutical composition as described above) to a subject.
- the present invention also provides a method for treating and/or preventing a disease in a subject which subject comprises cells of the invention, which method comprises the step of administering an agent to the subject wherein the agent is capable of controlling the release or expression of the dnSMAD or chimeric dnSMAD.
- this method involves administering an agent to a subject which already comprises cells of the present invention.
- the present methods for treating and/or preventing a disease may comprise administering an agent which increases the expression or activity of the dnSMAD or chimeric dnSMAD to a subject to which the engineered immune cell according to the present invention has been administered.
- a method for treating a disease relates to the therapeutic use of the cells of the present invention.
- the cells may be administered to a subject having an existing disease or condition in order to lessen, reduce or improve at least one symptom associated with the disease and/or to slow down, reduce or block the progression of the disease.
- the method may involve the steps of:
- nucleic acid construct according to the present invention introducing the nucleic acid construct according to the present invention, a first and second nucleic acid sequence as defined herein, a vector according to the present invention or a first and second vector as herein to the cell;
- the engineered immune effector cell may be administered in the form of a pharmaceutical composition.
- the pharmaceutical composition may additionally comprise a pharmaceutically acceptable carrier, diluent or excipient.
- the pharmaceutical composition may optionally comprise one or more further pharmaceutically active polypeptides and/or compounds.
- Such a formulation may, for example, be in a form suitable for intravenous infusion.
- the present invention provides a cell according to the present invention for use in treating a disease.
- the present invention also relates to the use of a cell according to the present invention for the manufacture of a medicament for the treatment of a disease.
- the disease to be treated and/or prevented by the method of the present invention may be cancer.
- the cancer may be a cancer such as neuroblastoma, multiple myeloma, prostate cancer, bladder cancer, breast cancer, colon cancer, endometrial cancer, kidney cancer (renal cell), leukaemia, lung cancer, melanoma, non-Hodgkin lymphoma, pancreatic cancer, and thyroid cancer.
- the cancer may be neuroblastoma.
- the cancer may be multiple myeloma.
- the cancer may be prostate cancer.
- the cell of the present invention may be capable of killing target cells, such as cancer cells.
- the target cell may be recognisable by expression of a TAA, for example the expression of a TAA provided above in Table 3.
- the cancer may be a cancer listed in Table 3.
- the administration of a cell according to the present invention can be accomplished using any of a variety of routes, such as intraperitoneally, intravenously, subcutaneously, transcutaneously or intramuscularly.
- Cells of the present invention may be generated by introducing DNA or RNA coding for dnSMAD or chimeric dnSMAD, as defined herein, by one of many means including transduction with a viral vector, transfection with DNA or RNA.
- the cell of the invention may be made by:
- the polynucleotide according to the present invention e.g. by transduction or transfection
- the nucleic acid construct or vector according to the present invention or a first and second nucleic acid sequence as defined herein, or a first and second vector as defined herein.
- the cell may be from a sample isolated from a subject.
- the present invention also provides a method of rendering an immune effector cell less susceptible to T ⁇ Rb by introducing a polynucleotide coding for a dnSMAD or chimeric dnSMAD to said immune effector cell.
- the method may comprise:
- the method of rendering an immune effector cell less susceptible to TQRb signalling may comprise maintaining the cell under conditions which allow the expression of the dnSMAD.
- the present invention further relates to the use of a dnSMAD to render an immune effector cell less susceptible to TQRb.
- Example 1 Investigating the capacity of truncated SMAD2 and truncated SMAD3 to block TGFp signalling in T cells
- T cells were generated expressing a second generation anti-GD2 CAR with a CD28-CD3z endodomain either alone or in combination with a truncated SMAD2 (dnSMAD2-MH2) or truncated SMAD3 (dnSMAD3-MH2) molecule.
- the cells were co-cultured with GD2-expressing SupT1 target cells at a 2:1 and 4:1 target: effector cell ratio.
- TGFp was spiked into the co-culture at 10ng/ml on day 0 and target cell killing was investigated at day 5 and 7 by flow cytometry. The results are shown in Figure 5.
- T cells expressing the CAR and dnSMAD2/SMAD3 or dnSMAD4 proteins (or CAR only) are depleted of CD56-expressing natural killer cells using the EasySep human CD56 positive selection kit (STEMCELL Technologies) according to the manufacturer’s instructions. Cells are then used in cytotoxicity assays after 1 , 5 and 7 days. Cytotoxicity assays are set up at a 1 :2 effector: target (E:T) cell ratio using SupT 1 expressing the target in presence/absence of human TGF-bI cells in 96-well plates. SupT1 WT cells are used in the same conditions as control. Not transduced T cells (NT T cells) are used in co-cultures with targets as a negative control. CAR-mediated cytotoxicity is assessed by flow cytometry after 1 , 5 and 7 days. T cells are identified from target cells by CD3 and staining. 7-AAD viability dye is used for exclusion of dead cells. T cell Proliferation Assay
- CTV staining is carried out.
- T cells expressing the different CAR constructs (NT T cells used as controls) are labelled with CTV before setup of co-cultures with target cells. Staining is performed by re-suspending the T cells at in fresh PBS containing CTV dye. Co-cultures are then set up with target cells expressing TGF-bI (and wild type cell as control), at an E:T ratio of 1 :2 and 1 :8 using. Proliferation is assessed by flow cytometry 5 and 7 days later.
- Cells are stained with 7-AAD and CD3 for exclusion of dead cells and detection of T cells, respectively, and the CTV-stained cells are used to measure proliferation by the extent of dye dilution of dead cells.
- Viable target cells are enumerated for each co-culture condition. The percentage of remaining target cells is calculated by normalizing the number of viable target cells of each condition to that recovered from co cultures carried out with NT T cells (100%).
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- General Health & Medical Sciences (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Genetics & Genomics (AREA)
- Zoology (AREA)
- Mycology (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Hematology (AREA)
- General Engineering & Computer Science (AREA)
- Oncology (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Biophysics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
GBGB1816399.8A GB201816399D0 (en) | 2018-10-08 | 2018-10-08 | Cell |
PCT/GB2019/052829 WO2020074868A1 (fr) | 2018-10-08 | 2019-10-07 | Cellule |
Publications (1)
Publication Number | Publication Date |
---|---|
EP3864034A1 true EP3864034A1 (fr) | 2021-08-18 |
Family
ID=64397609
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP19791327.0A Withdrawn EP3864034A1 (fr) | 2018-10-08 | 2019-10-07 | Cellule |
Country Status (3)
Country | Link |
---|---|
EP (1) | EP3864034A1 (fr) |
GB (1) | GB201816399D0 (fr) |
WO (1) | WO2020074868A1 (fr) |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN115181751A (zh) * | 2021-04-02 | 2022-10-14 | 苏州博腾生物制药有限公司 | 靶向白蛋白的嵌合抗原受体及其使用方法 |
-
2018
- 2018-10-08 GB GBGB1816399.8A patent/GB201816399D0/en not_active Ceased
-
2019
- 2019-10-07 WO PCT/GB2019/052829 patent/WO2020074868A1/fr unknown
- 2019-10-07 EP EP19791327.0A patent/EP3864034A1/fr not_active Withdrawn
Also Published As
Publication number | Publication date |
---|---|
WO2020074868A1 (fr) | 2020-04-16 |
GB201816399D0 (en) | 2018-11-28 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
EP3253783B1 (fr) | Système de signalisation | |
EP3185875B1 (fr) | Système de signalisation | |
EP3288969B1 (fr) | Construction d'acide nucléique pour l'expression de plusieurs recepteurs d'antigenes chimeriques. | |
US20240075066A1 (en) | Cell | |
JP2018532407A (ja) | 受容体 | |
KR20180041087A (ko) | 암을 치료하기 위한 방법 및 조성물 | |
CN111201033A (zh) | 细胞 | |
US20200338124A1 (en) | Cell | |
CN111479918A (zh) | 细胞 | |
WO2022029431A1 (fr) | Récepteur chimérique liant le tgf-bêta | |
US20240115606A1 (en) | Cell-Based Therapeutics Targeting CD70 | |
WO2017137759A1 (fr) | Système de signalisation | |
JP2024036532A (ja) | 操作された細胞溶解性免疫細胞 | |
EP3864034A1 (fr) | Cellule | |
US20210244762A1 (en) | Car-t cells expressing an inhibitory anti-tgf-beta-receptor ii single domain antibody | |
WO2022167798A1 (fr) | Molécule | |
CN115315439A (zh) | 细胞 | |
CN114555789A (zh) | 工程化免疫细胞 | |
WO2021123802A1 (fr) | Cellule | |
CN116194574A (zh) | 嵌合抗原受体细胞 | |
WO2019243817A1 (fr) | Cellule |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: UNKNOWN |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE |
|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE |
|
17P | Request for examination filed |
Effective date: 20210505 |
|
AK | Designated contracting states |
Kind code of ref document: A1 Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR |
|
DAV | Request for validation of the european patent (deleted) | ||
DAX | Request for extension of the european patent (deleted) | ||
RAP3 | Party data changed (applicant data changed or rights of an application transferred) |
Owner name: AUTOLUS LIMITED |
|
RIN1 | Information on inventor provided before grant (corrected) |
Inventor name: RIGHI, MATTEO Inventor name: DELLA PERUTA, MARCO Inventor name: THOMAS, SIMON Inventor name: ONUOHA, SHIMOBI Inventor name: CORDOBA, SHAUN Inventor name: PULE, MARTIN |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN |
|
18W | Application withdrawn |
Effective date: 20220324 |