NZ719720B2 - Engineered human t cell receptors with high affinity for wt1 - Google Patents
Engineered human t cell receptors with high affinity for wt1 Download PDFInfo
- Publication number
- NZ719720B2 NZ719720B2 NZ719720A NZ71972014A NZ719720B2 NZ 719720 B2 NZ719720 B2 NZ 719720B2 NZ 719720 A NZ719720 A NZ 719720A NZ 71972014 A NZ71972014 A NZ 71972014A NZ 719720 B2 NZ719720 B2 NZ 719720B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- tcr
- cell
- seq
- cells
- tcrs
- Prior art date
Links
- 108091008153 T cell receptors Proteins 0.000 title claims description 335
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 title claims description 335
- 241000282414 Homo sapiens Species 0.000 title claims description 33
- 239000000427 antigen Substances 0.000 claims abstract description 60
- 108091007172 antigens Proteins 0.000 claims abstract description 59
- 102000038129 antigens Human genes 0.000 claims abstract description 59
- 230000027455 binding Effects 0.000 claims description 77
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 63
- 102000026088 HLA-A2 Antigen Human genes 0.000 claims description 46
- 108010074032 HLA-A2 Antigen Proteins 0.000 claims description 46
- 201000011510 cancer Diseases 0.000 claims description 34
- 239000003814 drug Substances 0.000 claims description 27
- 238000004519 manufacturing process Methods 0.000 claims description 4
- 210000004027 cells Anatomy 0.000 abstract description 139
- 240000004808 Saccharomyces cerevisiae Species 0.000 abstract description 41
- 210000001744 T-Lymphocytes Anatomy 0.000 abstract description 40
- 230000001976 improved Effects 0.000 abstract description 10
- 230000000869 mutational Effects 0.000 abstract 1
- 208000008383 Wilms Tumor Diseases 0.000 description 122
- 102000004169 proteins and genes Human genes 0.000 description 61
- 108090000623 proteins and genes Proteins 0.000 description 61
- 235000018102 proteins Nutrition 0.000 description 58
- 102000004965 antibodies Human genes 0.000 description 47
- 108090001123 antibodies Proteins 0.000 description 47
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 40
- 230000014509 gene expression Effects 0.000 description 33
- 229920000023 polynucleotide Polymers 0.000 description 32
- 239000002157 polynucleotide Substances 0.000 description 32
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 32
- 235000001014 amino acid Nutrition 0.000 description 31
- 241000283707 Capra Species 0.000 description 26
- 150000001413 amino acids Chemical class 0.000 description 26
- 239000000203 mixture Substances 0.000 description 25
- 238000000034 method Methods 0.000 description 24
- 239000003446 ligand Substances 0.000 description 23
- 230000035772 mutation Effects 0.000 description 23
- 230000000694 effects Effects 0.000 description 21
- 229920003013 deoxyribonucleic acid Polymers 0.000 description 20
- 238000002818 protein evolution Methods 0.000 description 20
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 19
- 108090000765 processed proteins & peptides Proteins 0.000 description 19
- 102000004196 processed proteins & peptides Human genes 0.000 description 19
- 150000001875 compounds Chemical class 0.000 description 16
- 239000000539 dimer Substances 0.000 description 16
- 229920001850 Nucleic acid sequence Polymers 0.000 description 14
- 239000003795 chemical substances by application Substances 0.000 description 13
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 13
- 238000000338 in vitro Methods 0.000 description 13
- 230000001809 detectable Effects 0.000 description 12
- 229940079593 drugs Drugs 0.000 description 12
- 239000000243 solution Substances 0.000 description 12
- 230000001225 therapeutic Effects 0.000 description 12
- 241000588724 Escherichia coli Species 0.000 description 11
- 206010028980 Neoplasm Diseases 0.000 description 11
- 201000010099 disease Diseases 0.000 description 11
- -1 e.g. Substances 0.000 description 11
- 238000000684 flow cytometry Methods 0.000 description 11
- 239000000463 material Substances 0.000 description 11
- 229920001184 polypeptide Polymers 0.000 description 11
- 239000002246 antineoplastic agent Substances 0.000 description 10
- 238000010367 cloning Methods 0.000 description 10
- 238000006467 substitution reaction Methods 0.000 description 10
- 108010081208 RMFPNAPYL Proteins 0.000 description 9
- 108010004469 allophycocyanin Proteins 0.000 description 9
- 150000002500 ions Chemical class 0.000 description 9
- 150000007523 nucleic acids Chemical group 0.000 description 9
- 229920001405 Coding region Polymers 0.000 description 8
- 238000002823 phage display Methods 0.000 description 8
- 239000011780 sodium chloride Substances 0.000 description 8
- 230000000087 stabilizing Effects 0.000 description 8
- 239000000126 substance Substances 0.000 description 8
- 102100013077 CD4 Human genes 0.000 description 7
- 241000124008 Mammalia Species 0.000 description 7
- 229940014598 TAC Drugs 0.000 description 7
- 239000004480 active ingredient Substances 0.000 description 7
- 230000002068 genetic Effects 0.000 description 7
- 230000002401 inhibitory effect Effects 0.000 description 7
- 210000004962 mammalian cells Anatomy 0.000 description 7
- 238000002703 mutagenesis Methods 0.000 description 7
- 231100000350 mutagenesis Toxicity 0.000 description 7
- 108020004707 nucleic acids Proteins 0.000 description 7
- 239000000725 suspension Substances 0.000 description 7
- 230000001131 transforming Effects 0.000 description 7
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 238000004520 electroporation Methods 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 230000000670 limiting Effects 0.000 description 6
- 230000035800 maturation Effects 0.000 description 6
- 150000003839 salts Chemical class 0.000 description 6
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 5
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- 241000282412 Homo Species 0.000 description 5
- 241000235058 Komagataella pastoris Species 0.000 description 5
- 239000002775 capsule Substances 0.000 description 5
- 230000001472 cytotoxic Effects 0.000 description 5
- 231100000433 cytotoxic Toxicity 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 230000002708 enhancing Effects 0.000 description 5
- 238000009472 formulation Methods 0.000 description 5
- 230000012010 growth Effects 0.000 description 5
- 238000009396 hybridization Methods 0.000 description 5
- 230000003993 interaction Effects 0.000 description 5
- 239000002502 liposome Substances 0.000 description 5
- 230000004048 modification Effects 0.000 description 5
- 238000006011 modification reaction Methods 0.000 description 5
- 239000002773 nucleotide Substances 0.000 description 5
- 238000002360 preparation method Methods 0.000 description 5
- 230000011664 signaling Effects 0.000 description 5
- 238000010186 staining Methods 0.000 description 5
- 239000003053 toxin Substances 0.000 description 5
- 230000035897 transcription Effects 0.000 description 5
- 239000012099 Alexa Fluor family Substances 0.000 description 4
- 102100008857 MLANA Human genes 0.000 description 4
- 101710012533 MLANA Proteins 0.000 description 4
- 108020004999 Messenger RNA Proteins 0.000 description 4
- 108010070144 Single-Chain Antibodies Proteins 0.000 description 4
- 102000005632 Single-Chain Antibodies Human genes 0.000 description 4
- 231100000765 Toxin Toxicity 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- 238000007792 addition Methods 0.000 description 4
- 239000011324 bead Substances 0.000 description 4
- 230000003115 biocidal Effects 0.000 description 4
- 238000002659 cell therapy Methods 0.000 description 4
- 238000010276 construction Methods 0.000 description 4
- 239000008298 dragée Substances 0.000 description 4
- 230000001939 inductive effect Effects 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 238000005304 joining Methods 0.000 description 4
- 239000007788 liquid Substances 0.000 description 4
- 239000003550 marker Substances 0.000 description 4
- 229920002106 messenger RNA Polymers 0.000 description 4
- 125000003729 nucleotide group Chemical group 0.000 description 4
- 239000008194 pharmaceutical composition Substances 0.000 description 4
- 238000007747 plating Methods 0.000 description 4
- 230000001105 regulatory Effects 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 230000028327 secretion Effects 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 230000002588 toxic Effects 0.000 description 4
- 231100000331 toxic Toxicity 0.000 description 4
- 108020003112 toxins Proteins 0.000 description 4
- 230000002103 transcriptional Effects 0.000 description 4
- 229960005486 vaccines Drugs 0.000 description 4
- 235000014469 Bacillus subtilis Nutrition 0.000 description 3
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 3
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 3
- 108010053187 Diphtheria Toxin Proteins 0.000 description 3
- 102000016607 Diphtheria Toxin Human genes 0.000 description 3
- 241000196324 Embryophyta Species 0.000 description 3
- 108010010803 Gelatin Proteins 0.000 description 3
- 206010024324 Leukaemias Diseases 0.000 description 3
- 108010004729 Phycoerythrin Proteins 0.000 description 3
- 239000004698 Polyethylene (PE) Substances 0.000 description 3
- 108010026552 Proteome Proteins 0.000 description 3
- 108010039491 Ricin Proteins 0.000 description 3
- 229920002472 Starch Polymers 0.000 description 3
- 229940035295 Ting Drugs 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 230000000890 antigenic Effects 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 3
- 239000000969 carrier Substances 0.000 description 3
- 230000010261 cell growth Effects 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- 230000001419 dependent Effects 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 238000010586 diagram Methods 0.000 description 3
- 239000003937 drug carrier Substances 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 230000004927 fusion Effects 0.000 description 3
- 229920000159 gelatin Polymers 0.000 description 3
- 239000008273 gelatin Substances 0.000 description 3
- 235000019322 gelatine Nutrition 0.000 description 3
- 235000011852 gelatine desserts Nutrition 0.000 description 3
- 238000003384 imaging method Methods 0.000 description 3
- 230000003834 intracellular Effects 0.000 description 3
- 108010003486 leucyl-leucyl-phenylalanyl-glycyl-tyrosyl-prolyl-valyl-tyrosyl-valine Proteins 0.000 description 3
- 238000002826 magnetic-activated cell sorting Methods 0.000 description 3
- 239000000178 monomer Substances 0.000 description 3
- 229920001223 polyethylene glycol Polymers 0.000 description 3
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 108091007521 restriction endonucleases Proteins 0.000 description 3
- 239000011734 sodium Substances 0.000 description 3
- 229910052708 sodium Inorganic materials 0.000 description 3
- 239000000600 sorbitol Substances 0.000 description 3
- 241000894007 species Species 0.000 description 3
- 239000003381 stabilizer Substances 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 210000001519 tissues Anatomy 0.000 description 3
- 230000014621 translational initiation Effects 0.000 description 3
- 230000003612 virological Effects 0.000 description 3
- YJISHJVIRFPGGN-UHFFFAOYSA-N 5-[5-[3,4-dihydroxy-6-(hydroxymethyl)-5-methoxyoxan-2-yl]oxy-6-[[3,4-dihydroxy-6-(hydroxymethyl)-5-methoxyoxan-2-yl]oxymethyl]-3,4-dihydroxyoxan-2-yl]oxy-6-(hydroxymethyl)-2-methyloxane-3,4-diol Chemical compound O1C(CO)C(OC)C(O)C(O)C1OCC1C(OC2C(C(O)C(OC)C(CO)O2)O)C(O)C(O)C(OC2C(OC(C)C(O)C2O)CO)O1 YJISHJVIRFPGGN-UHFFFAOYSA-N 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N ADRIAMYCIN Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 2
- 240000000800 Allium ursinum Species 0.000 description 2
- NWMHDZMRVUOQGL-CZEIJOLGSA-N Almurtide Chemical compound OC(=O)CC[C@H](C(N)=O)NC(=O)[C@H](C)NC(=O)CO[C@@H]([C@H](O)[C@H](O)CO)[C@@H](NC(C)=O)C=O NWMHDZMRVUOQGL-CZEIJOLGSA-N 0.000 description 2
- 241000228212 Aspergillus Species 0.000 description 2
- 241000193830 Bacillus <bacterium> Species 0.000 description 2
- 241000894006 Bacteria Species 0.000 description 2
- 206010006187 Breast cancer Diseases 0.000 description 2
- 241000701489 Cauliflower mosaic virus Species 0.000 description 2
- 210000000170 Cell Membrane Anatomy 0.000 description 2
- 108020004705 Codon Proteins 0.000 description 2
- 206010009944 Colon cancer Diseases 0.000 description 2
- 229920002676 Complementary DNA Polymers 0.000 description 2
- 241000699802 Cricetulus griseus Species 0.000 description 2
- GZCGUPFRVQAUEE-KCDKBNATSA-N D-(+)-Galactose Natural products OC[C@@H](O)[C@H](O)[C@H](O)[C@@H](O)C=O GZCGUPFRVQAUEE-KCDKBNATSA-N 0.000 description 2
- 229960004679 Doxorubicin Drugs 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- 241000283073 Equus caballus Species 0.000 description 2
- 240000007842 Glycine max Species 0.000 description 2
- 235000010469 Glycine max Nutrition 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N HCl Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- 240000005979 Hordeum vulgare Species 0.000 description 2
- 235000007340 Hordeum vulgare Nutrition 0.000 description 2
- 102000018358 Immunoglobulins Human genes 0.000 description 2
- 108060003951 Immunoglobulins Proteins 0.000 description 2
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 2
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 2
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 2
- GUBGYTABKSRVRQ-UUNJERMWSA-N Lactose Natural products O([C@@H]1[C@H](O)[C@H](O)[C@H](O)O[C@@H]1CO)[C@H]1[C@@H](O)[C@@H](O)[C@H](O)[C@H](CO)O1 GUBGYTABKSRVRQ-UUNJERMWSA-N 0.000 description 2
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 2
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 2
- 206010033128 Ovarian cancer Diseases 0.000 description 2
- 210000001672 Ovary Anatomy 0.000 description 2
- 108010077397 Pseudomonas aeruginosa toxA protein Proteins 0.000 description 2
- 108020004511 Recombinant DNA Proteins 0.000 description 2
- 206010038389 Renal cancer Diseases 0.000 description 2
- 241000235403 Rhizomucor miehei Species 0.000 description 2
- 235000003534 Saccharomyces carlsbergensis Nutrition 0.000 description 2
- 229940081969 Saccharomyces cerevisiae Drugs 0.000 description 2
- 235000007238 Secale cereale Nutrition 0.000 description 2
- 240000002057 Secale cereale Species 0.000 description 2
- YYGNTYWPHWGJRM-RUSDCZJESA-N Squalene Natural products C(=C\CC/C(=C\CC/C=C(\CC/C=C(\CC/C=C(\C)/C)/C)/C)/C)(\CC/C=C(\C)/C)/C YYGNTYWPHWGJRM-RUSDCZJESA-N 0.000 description 2
- 108010090804 Streptavidin Proteins 0.000 description 2
- 206010057644 Testis cancer Diseases 0.000 description 2
- 229920001949 Transfer RNA Polymers 0.000 description 2
- 241000499912 Trichoderma reesei Species 0.000 description 2
- 240000008529 Triticum aestivum Species 0.000 description 2
- 230000035531 V beta Effects 0.000 description 2
- 240000008042 Zea mays Species 0.000 description 2
- 235000002017 Zea mays subsp mays Nutrition 0.000 description 2
- QTBSBXVTEAMEQO-UHFFFAOYSA-N acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 2
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 230000000240 adjuvant Effects 0.000 description 2
- 125000003277 amino group Chemical group 0.000 description 2
- 239000000611 antibody drug conjugate Substances 0.000 description 2
- 108091008116 antibody drug conjugates Proteins 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- 239000001768 carboxy methyl cellulose Substances 0.000 description 2
- 239000001913 cellulose Substances 0.000 description 2
- 229920002678 cellulose Polymers 0.000 description 2
- 230000000295 complement Effects 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 235000005822 corn Nutrition 0.000 description 2
- 235000005824 corn Nutrition 0.000 description 2
- 230000000875 corresponding Effects 0.000 description 2
- 230000001186 cumulative Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 239000010685 fatty oil Substances 0.000 description 2
- 239000000945 filler Substances 0.000 description 2
- 238000003260 fluorescence intensity Methods 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 238000002744 homologous recombination Methods 0.000 description 2
- 200000000003 influenza A Diseases 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 230000000977 initiatory Effects 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 201000010982 kidney cancer Diseases 0.000 description 2
- 239000008101 lactose Substances 0.000 description 2
- GUBGYTABKSRVRQ-XLOQQCSPSA-N lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 201000007270 liver cancer Diseases 0.000 description 2
- 201000005202 lung cancer Diseases 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 239000006249 magnetic particle Substances 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 230000003211 malignant Effects 0.000 description 2
- 230000001404 mediated Effects 0.000 description 2
- 229960000485 methotrexate Drugs 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 238000003032 molecular docking Methods 0.000 description 2
- 108010045030 monoclonal antibodies Proteins 0.000 description 2
- 102000005614 monoclonal antibodies Human genes 0.000 description 2
- 230000001613 neoplastic Effects 0.000 description 2
- 201000008026 nephroblastoma Diseases 0.000 description 2
- 230000001264 neutralization Effects 0.000 description 2
- MUBZPKHOEPUJKR-UHFFFAOYSA-N oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 2
- 239000002245 particle Substances 0.000 description 2
- 239000000825 pharmaceutical preparation Substances 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 2
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 2
- 210000001236 prokaryotic cell Anatomy 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 210000001938 protoplasts Anatomy 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 229920002973 ribosomal RNA Polymers 0.000 description 2
- 201000000849 skin cancer Diseases 0.000 description 2
- KEAYESYHFKHZAL-UHFFFAOYSA-N sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 2
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 2
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 2
- 235000019698 starch Nutrition 0.000 description 2
- 239000007858 starting material Substances 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 235000000346 sugar Nutrition 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 230000002194 synthesizing Effects 0.000 description 2
- 238000010189 synthetic method Methods 0.000 description 2
- 239000000454 talc Substances 0.000 description 2
- 229910052623 talc Inorganic materials 0.000 description 2
- 201000003120 testicular cancer Diseases 0.000 description 2
- GWEVSGVZZGPLCZ-UHFFFAOYSA-N titan oxide Chemical compound O=[Ti]=O GWEVSGVZZGPLCZ-UHFFFAOYSA-N 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 238000004450 types of analysis Methods 0.000 description 2
- 238000011144 upstream manufacturing Methods 0.000 description 2
- 235000021307 wheat Nutrition 0.000 description 2
- MIJDSYMOBYNHOT-UHFFFAOYSA-N 2-(ethylamino)ethanol Chemical compound CCNCCO MIJDSYMOBYNHOT-UHFFFAOYSA-N 0.000 description 1
- HZLCGUXUOFWCCN-UHFFFAOYSA-N 2-hydroxynonadecane-1,2,3-tricarboxylic acid Chemical compound CCCCCCCCCCCCCCCCC(C(O)=O)C(O)(C(O)=O)CC(O)=O HZLCGUXUOFWCCN-UHFFFAOYSA-N 0.000 description 1
- GHASVSINZRGABV-UHFFFAOYSA-N 5-flurouricil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 1
- 101700039609 AGA2 Proteins 0.000 description 1
- 101710034857 ATIC Proteins 0.000 description 1
- 108010066676 Abrin Proteins 0.000 description 1
- 244000215068 Acacia senegal Species 0.000 description 1
- 102000013563 Acid Phosphatase Human genes 0.000 description 1
- 108010051457 Acid Phosphatase Proteins 0.000 description 1
- 229940009456 Adriamycin Drugs 0.000 description 1
- 239000012114 Alexa Fluor 647 Substances 0.000 description 1
- XJKJWTWGDGIQRH-BFIDDRIFSA-N Alginic acid Chemical compound O1[C@@H](C(O)=O)[C@@H](OC)[C@H](O)[C@H](O)[C@@H]1O[C@@H]1[C@@H](C(O)=O)O[C@@H](C)[C@@H](O)[C@H]1O XJKJWTWGDGIQRH-BFIDDRIFSA-N 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K Aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- AVKUERGKIZMTKX-NJBDSQKTSA-N Ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 1
- 229940064005 Antibiotic throat preparations Drugs 0.000 description 1
- 229940083879 Antibiotics FOR TREATMENT OF HEMORRHOIDS AND ANAL FISSURES FOR TOPICAL USE Drugs 0.000 description 1
- 229940042052 Antibiotics for systemic use Drugs 0.000 description 1
- 244000105975 Antidesma platyphyllum Species 0.000 description 1
- 102000006306 Antigen Receptors Human genes 0.000 description 1
- 108010083359 Antigen Receptors Proteins 0.000 description 1
- 229940042786 Antitubercular Antibiotics Drugs 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 240000003291 Armoracia rusticana Species 0.000 description 1
- 235000011330 Armoracia rusticana Nutrition 0.000 description 1
- 229960001230 Asparagine Drugs 0.000 description 1
- 229960005261 Aspartic Acid Drugs 0.000 description 1
- 241001513093 Aspergillus awamori Species 0.000 description 1
- 241000228195 Aspergillus ficuum Species 0.000 description 1
- 241000228245 Aspergillus niger Species 0.000 description 1
- 241000416162 Astragalus gummifer Species 0.000 description 1
- 241000194108 Bacillus licheniformis Species 0.000 description 1
- 240000008371 Bacillus subtilis Species 0.000 description 1
- 229940075615 Bacillus subtilis Drugs 0.000 description 1
- 108010071919 Bispecific Antibodies Proteins 0.000 description 1
- 210000001124 Body Fluids Anatomy 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 235000006008 Brassica napus var napus Nutrition 0.000 description 1
- 240000000385 Brassica napus var. napus Species 0.000 description 1
- 235000006618 Brassica rapa subsp oleifera Nutrition 0.000 description 1
- 235000004977 Brassica sinapistrum Nutrition 0.000 description 1
- 210000001266 CD8-Positive T-Lymphocytes Anatomy 0.000 description 1
- 102000004040 Capsid Proteins Human genes 0.000 description 1
- 108090000565 Capsid Proteins Proteins 0.000 description 1
- 210000004520 Cell Wall Skeleton Anatomy 0.000 description 1
- 108010039939 Cell Wall Skeleton Proteins 0.000 description 1
- 210000003578 Chromosomes, Bacterial Anatomy 0.000 description 1
- 230000037250 Clearance Effects 0.000 description 1
- 229920000453 Consensus sequence Polymers 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 239000004971 Cross linker Substances 0.000 description 1
- 210000000805 Cytoplasm Anatomy 0.000 description 1
- MTCFGRXMJLQNBG-UWTATZPHSA-N D-serine Chemical compound OC[C@@H](N)C(O)=O MTCFGRXMJLQNBG-UWTATZPHSA-N 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N D-sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- QIVBCDIJIAJPQS-SECBINFHSA-N D-tryptophane Chemical compound C1=CC=C2C(C[C@@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-SECBINFHSA-N 0.000 description 1
- STQGQHZAVUOBTE-VGBVRHCVSA-N DAUNOMYCIN Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 1
- 238000007399 DNA isolation Methods 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 101700011961 DPOM Proteins 0.000 description 1
- 229960000975 Daunorubicin Drugs 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 1
- VQNATVDKACXKTF-XELLLNAOSA-N Duocarmycin Chemical compound COC1=C(OC)C(OC)=C2NC(C(=O)N3C4=CC(=O)C5=C([C@@]64C[C@@H]6C3)C=C(N5)C(=O)OC)=CC2=C1 VQNATVDKACXKTF-XELLLNAOSA-N 0.000 description 1
- 241000448280 Elates Species 0.000 description 1
- 210000003743 Erythrocytes Anatomy 0.000 description 1
- 229940023064 Escherichia coli Drugs 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N Ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 102100009142 FANCC Human genes 0.000 description 1
- 101700023762 FANCC Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 108010067306 Fibronectins Proteins 0.000 description 1
- 102000016359 Fibronectins Human genes 0.000 description 1
- 229960002949 Fluorouracil Drugs 0.000 description 1
- 102200036988 GALNT4 D51G Human genes 0.000 description 1
- 206010017758 Gastric cancer Diseases 0.000 description 1
- 229960002989 Glutamic Acid Drugs 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 229920000084 Gum arabic Polymers 0.000 description 1
- 229940093922 Gynecological Antibiotics Drugs 0.000 description 1
- 101700049607 HIS4 Proteins 0.000 description 1
- 206010073071 Hepatocellular carcinoma Diseases 0.000 description 1
- 102000006755 Human Immunodeficiency Virus gag Gene Products Human genes 0.000 description 1
- 108010086541 Human Immunodeficiency Virus gag Gene Products Proteins 0.000 description 1
- 210000004408 Hybridomas Anatomy 0.000 description 1
- 229960000310 ISOLEUCINE Drugs 0.000 description 1
- 102000009786 Immunoglobulin Constant Regions Human genes 0.000 description 1
- 108010009817 Immunoglobulin Constant Regions Proteins 0.000 description 1
- 210000003000 Inclusion Bodies Anatomy 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N Isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- SBUJHOSQTJFQJX-NOAMYHISSA-N Kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 description 1
- 241001138401 Kluyveromyces lactis Species 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-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
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-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
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-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
- 125000000510 L-tryptophano group Chemical group [H]C1=C([H])C([H])=C2N([H])C([H])=C(C([H])([H])[C@@]([H])(C(O[H])=O)N([H])[*])C2=C1[H] 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 101710029649 MDV043 Proteins 0.000 description 1
- 235000019759 Maize starch Nutrition 0.000 description 1
- 206010025650 Malignant melanoma Diseases 0.000 description 1
- 241001599018 Melanogaster Species 0.000 description 1
- FEWJPZIEWOKRBE-XIXRPRMCSA-N Mesotartaric acid Chemical compound OC(=O)[C@@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-XIXRPRMCSA-N 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 229960004452 Methionine Drugs 0.000 description 1
- 229960004857 Mitomycin Drugs 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 1
- 101710008008 NEUROD6 Proteins 0.000 description 1
- 102100015077 NEUROD6 Human genes 0.000 description 1
- 101700080605 NUC1 Proteins 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 208000002154 Non-Small-Cell Lung Carcinoma Diseases 0.000 description 1
- 206010025310 Other lymphomas Diseases 0.000 description 1
- 101710021097 PAPSS1 Proteins 0.000 description 1
- 101700061424 POLB Proteins 0.000 description 1
- 229960005190 Phenylalanine Drugs 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 235000002343 Primula veris Nutrition 0.000 description 1
- 240000003122 Primula vulgaris Species 0.000 description 1
- 235000016311 Primula vulgaris Nutrition 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N Procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960002429 Proline Drugs 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 210000003324 RBC Anatomy 0.000 description 1
- 101700054624 RF1 Proteins 0.000 description 1
- 229940100486 RICE STARCH Drugs 0.000 description 1
- 101700079635 RTL1 Proteins 0.000 description 1
- 229920001914 Ribonucleotide Polymers 0.000 description 1
- 240000000528 Ricinus communis Species 0.000 description 1
- 235000004443 Ricinus communis Nutrition 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 101700062671 SIAE Proteins 0.000 description 1
- 101700018407 SPHK1 Proteins 0.000 description 1
- 102100005042 SPHK1 Human genes 0.000 description 1
- 210000002966 Serum Anatomy 0.000 description 1
- 241000580858 Simian-Human immunodeficiency virus Species 0.000 description 1
- 240000002967 Sium sisarum Species 0.000 description 1
- 208000000587 Small Cell Lung Carcinoma Diseases 0.000 description 1
- 240000001016 Solanum tuberosum Species 0.000 description 1
- 235000002595 Solanum tuberosum Nutrition 0.000 description 1
- 229940031439 Squalene Drugs 0.000 description 1
- CZMRCDWAGMRECN-GDQSFJPYSA-N Sucrose Natural products O([C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@H](CO)O1)[C@@]1(CO)[C@H](O)[C@@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-GDQSFJPYSA-N 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- 210000003283 T-Lymphocytes, Helper-Inducer Anatomy 0.000 description 1
- 108060008443 TPPP Proteins 0.000 description 1
- MPLHNVLQVRSVEE-UHFFFAOYSA-N Texas Red Chemical compound [O-]S(=O)(=O)C1=CC(S(Cl)(=O)=O)=CC=C1C(C1=CC=2CCCN3CCCC(C=23)=C1O1)=C2C1=C(CCC1)C3=[N+]1CCCC3=C2 MPLHNVLQVRSVEE-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 210000001541 Thymus Gland Anatomy 0.000 description 1
- 229940024982 Topical Antifungal Antibiotics Drugs 0.000 description 1
- 229920001615 Tragacanth Polymers 0.000 description 1
- GETQZCLCWQTVFV-UHFFFAOYSA-N Trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 1
- HRXKRNGNAMMEHJ-UHFFFAOYSA-K Trisodium citrate Chemical compound [Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O HRXKRNGNAMMEHJ-UHFFFAOYSA-K 0.000 description 1
- 229960004799 Tryptophan Drugs 0.000 description 1
- UGGWPQSBPIFKDZ-KOTLKJBCSA-N Vindesine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(N)=O)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1N=C1[C]2C=CC=C1 UGGWPQSBPIFKDZ-KOTLKJBCSA-N 0.000 description 1
- 229960004355 Vindesine Drugs 0.000 description 1
- 102000016350 Viral Proteins Human genes 0.000 description 1
- 108010067390 Viral Proteins Proteins 0.000 description 1
- 229940100445 WHEAT STARCH Drugs 0.000 description 1
- JMUHBNWAORSSBD-WKYWBUFDSA-N [(2R)-3-[2-[[(2S)-2-[[(4R)-4-[[(2S)-2-[[(2R)-2-[(3R,4R,5S,6R)-3-acetamido-2,5-dihydroxy-6-(hydroxymethyl)oxan-4-yl]oxypropanoyl]amino]propanoyl]amino]-5-amino-5-oxopentanoyl]amino]propanoyl]amino]ethoxy-hydroxyphosphoryl]oxy-2-hexadecanoyloxypropyl] hexad Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@@H](OC(=O)CCCCCCCCCCCCCCC)COP(O)(=O)OCCNC(=O)[C@H](C)NC(=O)CC[C@H](C(N)=O)NC(=O)[C@H](C)NC(=O)[C@@H](C)O[C@H]1[C@H](O)[C@@H](CO)OC(O)[C@@H]1NC(C)=O JMUHBNWAORSSBD-WKYWBUFDSA-N 0.000 description 1
- 235000010489 acacia gum Nutrition 0.000 description 1
- 239000000205 acacia gum Substances 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000002378 acidificating Effects 0.000 description 1
- NIXOWILDQLNWCW-UHFFFAOYSA-N acrylic acid Chemical compound OC(=O)C=C NIXOWILDQLNWCW-UHFFFAOYSA-N 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 229940040563 agaric acid Drugs 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 239000004037 angiogenesis inhibitor Substances 0.000 description 1
- 229940121369 angiogenesis inhibitors Drugs 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000003432 anti-folate Effects 0.000 description 1
- 230000003110 anti-inflammatory Effects 0.000 description 1
- 229960000070 antineoplastic Monoclonal antibodies Drugs 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 101700029951 aroK Proteins 0.000 description 1
- 101700045673 aroK1 Proteins 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 235000020054 awamori Nutrition 0.000 description 1
- 238000002869 basic local alignment search tool Methods 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 238000004166 bioassay Methods 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 239000012472 biological sample Substances 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 210000003969 blast cell Anatomy 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 235000008984 brauner Senf Nutrition 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000001413 cellular Effects 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 238000005352 clarification Methods 0.000 description 1
- 230000035512 clearance Effects 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 201000011231 colorectal cancer Diseases 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 210000004748 cultured cells Anatomy 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 230000001086 cytosolic Effects 0.000 description 1
- 230000003111 delayed Effects 0.000 description 1
- 238000004925 denaturation Methods 0.000 description 1
- 230000036425 denaturation Effects 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 238000002059 diagnostic imaging Methods 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 230000003292 diminished Effects 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 230000001804 emulsifying Effects 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 238000006911 enzymatic reaction Methods 0.000 description 1
- 238000001976 enzyme digestion Methods 0.000 description 1
- 239000002532 enzyme inhibitor Substances 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 201000005619 esophageal carcinoma Diseases 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 230000037320 fibronectin Effects 0.000 description 1
- 238000011049 filling Methods 0.000 description 1
- 125000003983 fluorenyl group Chemical group C1(=CC=CC=2C3=CC=CC=C3CC12)* 0.000 description 1
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 1
- 238000000799 fluorescence microscopy Methods 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 239000004052 folic acid antagonist Substances 0.000 description 1
- 230000037406 food intake Effects 0.000 description 1
- 238000005755 formation reaction Methods 0.000 description 1
- VZCYOOQTPOCHFL-UHFFFAOYSA-N fumaric acid Chemical compound OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037240 fusion proteins Human genes 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 235000004554 glutamine Nutrition 0.000 description 1
- 230000003899 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 238000000227 grinding Methods 0.000 description 1
- 235000009424 haa Nutrition 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 230000002209 hydrophobic Effects 0.000 description 1
- 150000004679 hydroxides Chemical class 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000000415 inactivating Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 230000000968 intestinal Effects 0.000 description 1
- 229940079866 intestinal antibiotics Drugs 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- XEEYBQQBJWHFJM-UHFFFAOYSA-N iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- FZWBNHMXJMCXLU-BLAUPYHCSA-N isomaltotriose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1OC[C@@H]1[C@@H](O)[C@H](O)[C@@H](O)[C@@H](OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C=O)O1 FZWBNHMXJMCXLU-BLAUPYHCSA-N 0.000 description 1
- 229960000318 kanamycin Drugs 0.000 description 1
- 230000002147 killing Effects 0.000 description 1
- 239000004922 lacquer Substances 0.000 description 1
- 210000002429 large intestine Anatomy 0.000 description 1
- 229960003136 leucine Drugs 0.000 description 1
- 101710030587 ligN Proteins 0.000 description 1
- 101700077585 ligd Proteins 0.000 description 1
- GZQKNULLWNGMCW-PWQABINMSA-J lipid A(4-) Chemical compound O1[C@H](CO)[C@@H](OP([O-])([O-])=O)[C@H](OC(=O)C[C@@H](CCCCCCCCCCC)OC(=O)CCCCCCCCCCCCC)[C@@H](NC(=O)C[C@@H](CCCCCCCCCCC)OC(=O)CCCCCCCCCCC)[C@@H]1OC[C@@H]1[C@@H](O)[C@H](OC(=O)C[C@H](O)CCCCCCCCCCC)[C@@H](NC(=O)C[C@H](O)CCCCCCCCCCC)[C@@H](OP([O-])([O-])=O)O1 GZQKNULLWNGMCW-PWQABINMSA-J 0.000 description 1
- 229940057995 liquid paraffin Drugs 0.000 description 1
- 238000005567 liquid scintillation counting Methods 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 239000006194 liquid suspension Substances 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 101710028189 lsc Proteins 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 201000005244 lung non-small cell carcinoma Diseases 0.000 description 1
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 239000011976 maleic acid Substances 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 125000001160 methoxycarbonyl group Chemical group [H]C([H])([H])OC(*)=O 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 239000011325 microbead Substances 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 229960005225 mifamurtide Drugs 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 229960000060 monoclonal antibodies Drugs 0.000 description 1
- 238000002887 multiple sequence alignment Methods 0.000 description 1
- 230000017066 negative regulation of growth Effects 0.000 description 1
- 101700006494 nucA Proteins 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 238000007899 nucleic acid hybridization Methods 0.000 description 1
- 239000003865 nucleic acid synthesis inhibitor Substances 0.000 description 1
- 229940005935 ophthalmologic Antibiotics Drugs 0.000 description 1
- 230000004768 organ dysfunction Effects 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 239000003791 organic solvent mixture Substances 0.000 description 1
- 210000000056 organs Anatomy 0.000 description 1
- 201000008968 osteosarcoma Diseases 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 239000006179 pH buffering agent Substances 0.000 description 1
- 238000004091 panning Methods 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000002093 peripheral Effects 0.000 description 1
- 239000000546 pharmaceutic aid Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 235000011007 phosphoric acid Nutrition 0.000 description 1
- 150000003016 phosphoric acids Chemical class 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000000865 phosphorylative Effects 0.000 description 1
- 239000000049 pigment Substances 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 239000004014 plasticizer Substances 0.000 description 1
- 108010078110 pokeweed antiviral protein Proteins 0.000 description 1
- 229920001888 polyacrylic acid Polymers 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 229920001592 potato starch Polymers 0.000 description 1
- 230000003389 potentiating Effects 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 229940002612 prodrugs Drugs 0.000 description 1
- 230000004853 protein function Effects 0.000 description 1
- 238000001742 protein purification Methods 0.000 description 1
- 239000012460 protein solution Substances 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 230000002285 radioactive Effects 0.000 description 1
- 239000002464 receptor antagonist Substances 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 238000000611 regression analysis Methods 0.000 description 1
- 230000022983 regulation of cell cycle Effects 0.000 description 1
- 230000001177 retroviral Effects 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 239000011435 rock Substances 0.000 description 1
- 101700057137 sacB Proteins 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 238000007480 sanger sequencing Methods 0.000 description 1
- 238000007423 screening assay Methods 0.000 description 1
- 230000003248 secreting Effects 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 239000002002 slurry Substances 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 239000001509 sodium citrate Substances 0.000 description 1
- 239000007901 soft capsule Substances 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000012439 solid excipient Substances 0.000 description 1
- 230000000392 somatic Effects 0.000 description 1
- 230000003393 splenic Effects 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 230000004936 stimulating Effects 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 239000004575 stone Substances 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 238000005670 sulfation reaction Methods 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 235000012222 talc Nutrition 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 229960001367 tartaric acid Drugs 0.000 description 1
- 102000015610 tax Gene Products Human genes 0.000 description 1
- 108010038751 tax Gene Products Proteins 0.000 description 1
- 230000002992 thymic Effects 0.000 description 1
- 239000004408 titanium dioxide Substances 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 230000024664 tolerance induction Effects 0.000 description 1
- 231100000167 toxic agent Toxicity 0.000 description 1
- 102000003995 transcription factors Human genes 0.000 description 1
- 108090000464 transcription factors Proteins 0.000 description 1
- LXZZYRPGZAFOLE-UHFFFAOYSA-L transplatin Chemical compound [H][N]([H])([H])[Pt](Cl)(Cl)[N]([H])([H])[H] LXZZYRPGZAFOLE-UHFFFAOYSA-L 0.000 description 1
- XETCRXVKJHBPMK-MJSODCSWSA-N trehalose 6,6'-dimycolate Chemical compound C([C@@H]1[C@H]([C@H](O)[C@@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](COC(=O)C(CCCCCCCCCCC3C(C3)CCCCCCCCCCCCCCCCCC)C(O)CCCCCCCCCCCCCCCCCCCCCCCCC)O2)O)O1)O)OC(=O)C(C(O)CCCCCCCCCCCCCCCCCCCCCCCCC)CCCCCCCCCCC1CC1CCCCCCCCCCCCCCCCCC XETCRXVKJHBPMK-MJSODCSWSA-N 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- 239000011778 trisodium citrate Substances 0.000 description 1
- 230000001228 trophic Effects 0.000 description 1
- 210000004881 tumor cells Anatomy 0.000 description 1
- 108010087967 type I signal peptidase Proteins 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 229960004295 valine Drugs 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N β-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
- A61K35/17—Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/177—Receptors; Cell surface antigens; Cell surface determinants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/177—Receptors; Cell surface antigens; Cell surface determinants
- A61K38/1774—Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4747—Apoptosis related proteins
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/565—Complementarity determining region [CDR]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/60—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
- C07K2317/62—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
- C07K2317/622—Single chain antibody (scFv)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
-
- C—CHEMISTRY; METALLURGY
- 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
Abstract
cell receptors (TCRs) that have specificity for the WT1 antigen are provided. The TCRs include higher affinity TCRs that were engineered through the generation of mutational libraries of TCRs in a single-chain format, followed by selection for improved stability and affinity on the surface of yeast (i.e. directed evolution). In embodiments, the TCRs can be used in soluble form for targeted delivery in vivo, or as genes introduced into T cells in an adoptive T cell setting. t (i.e. directed evolution). In embodiments, the TCRs can be used in soluble form for targeted delivery in vivo, or as genes introduced into T cells in an adoptive T cell setting.
Description
ENGINEERED HUMAN T CELL ORS WITH HIGH TY FOR WT1
CROSS-REFERENCE TO D APPLICATIONS
This application claims the benefit under 35 U.S.C. § 119(e) of U.S.
Provisional Patent Application No. 61/907,887 filed November 22, 2013, and this
provisional application is incorporated herein by reference in its entirety.
STATEMENT REGUARDING FEDERALLY SPONSORED CH OR
DEVELOPMENT
This disclosure was made with U.S. Government support under Grant
numbers R01 GM55767 and T32 GM070421, awarded by the National Institutes of
Health. The U.S. Government has certain rights in the disclosure.
STATEMENT REGARDING SEQUENCE LISTING
The Sequence g associated with this application is provided in text
format in lieu of a paper copy, and is hereby incorporated by reference into the
specification. The name of the text file ning the Sequence Listing is
IMMU_003_02WO_ST25.txt. The text file is 18 KB, was created on November 21,
2014 and is being submitted electronically via b.
FIELD OF THE INVENTION
The disclosure relates to T cell receptors (TCR) against the Wilms' tumor
antigen (WT1), ing high-affinity TCRs engineered by in vitro techniques, as
well as methods of producing modified TCRs and single-chain TCRs and the
ponding uses of the TCRs for therapeutic, diagnostic, and imaging methods.
BACKGROUND
T cell receptors (TCRs) and antibodies are molecules that have evolved to
recognize different classes of antigens (ligands)((Murphy (2012), xix, 868 p.)). TCRs
are antigen-specific molecules that are responsible for recognizing antigenic
peptides presented in the context of a product of the major histocompatibility
complex (MHC) on the surface of antigen ting cells (APCs) or any nucleated
cell (e.g., all human cells in the body, except red blood cells). In contrast, antibodies
typically recognize soluble or cell-surface ns, and do not require presentation
of the antigen by an MHC. This system endows T cells, via their TCRs, with the
potential ability to ize the entire array of intracellular antigens sed by a
cell (including virus ns) that are processed intracellularly into short peptides,
bound to an intracellular MHC molecule, and delivered to the surface as a peptide-
MHC complex (pepMHC). This system allows virtually any foreign protein (e.g.,
mutated cancer antigen or virus protein) or aberrantly expressed protein to serve a
target for T cells (reviewed in (Davis and Bjorkman (1988) Nature, 334, 395—402;
Davis et al. (1998) Annu Rev Immunol, 16, 523—544; Murphy (2012), xix, 868 p.)).
The interaction of a TCR and a pepMHC can drive the T cell into various
states of activation, depending on the affinity (or dissociation rate) of binding. The
TCR recognition process allows a T cell to discriminate between a normal, healthy
cell and, e.g., one that has become transformed via a virus or malignancy, by
providing a diverse repertoire of TCRs, wherein there is a high probability that one or
more TCRs will be t with a binding ty for the foreign peptide bound to an
MHC molecule that is above the threshold for stimulating T cell activity (Manning and
Kranz (1999) logy Today, 20, 417-422).
To date, wild type TCRs isolated from either human or mouse T cell clones
that were identified by in vitro culturing have been shown to have relatively low
binding affinities (Kd = 1 — 300 uM) (Davis et al. (1998) Annu Rev Immunol, 16, 523-
544). Part of the explanation for this seems to be that T cells that develop in the
thymus are negatively ed (tolerance induction) on self-pepMHC ligands, such
that T cells with too high of an affinity are deleted (Starr et al. (2003) Annu Rev
Immunol, 21, 139-76). To compensate for these relatively low affinities, T cells have
evolved a co-receptor system in which the cell surface molecules CD4 and CD8 bind
to the MHC les (class II and class I, tively) and ize with the TCR
in mediating signaling activity. CD8 is particularly effective in this process, allowing
TCRs with very low affinity (e.g.,Kd =300 uM) to mediate potent antigen-specific
activity.
In vitro, directed evolution has been used to generate TCRs with higher
affinity for a specific pepMHC. The three different display methods that have been
used are yeast display (Holler et al. (2003) Nat Immunol, 4, 55-62; Holler et al.
(2000) Proc Natl Acad Sci U S A, 97, 5387-92), phage display (Li et al. (2005) Nat
Biotechnol, 23, 349-54), and T cell display (Chervin et al. (2008) J Immunol s,
339, 175-84). In all three approaches, the process involves engineering, or
modifying, a TCR that ts the normal, low affinity of the wild—type TCR, so that
affinity of s of the TCR have increased affinity for the cognate pepMHC (the
original antigen that the T cells were specific for). Thus, the wild-type TCR was used
as a template for producing mutagenized libraries in one or more of the CDRs, and
mutants with higher affinity were selected by binding to the cognate peptide-MHC
anfigen.
In the present disclosure, a wild-type T cell receptor and high affinity T cell
receptors specific for the Wilm’s Tumor-1 (WT1) engineered by yeast y are
disclosed. WT1 is a transcription factor that has been described to function both as
a tumor suppressor and an oncogene. WT1 is expressed at high levels in leukemia
as well as a variety of solid tumors (Sugiyama et al. (2010) se Journal of
Clinical gy 40(5) 377-387). It has been the target of vaccine efforts, and
various adoptive T cell approaches using T cells with wild-type T cell receptors.
WT1 has been ranked number 1 in a prioritization list of the top 75 cancer
antigens by the National Cancer Institute er et al. (2009) Clin Cancer Res, 15,
5323—5337). Accordingly, there is a need to identify agents, e.g., therapeutic agents,
that specifically target this cancer n. The present ion provides in vitro
engineered, higher affinity TCRs that can be used, e.g., in soluble form for targeted
delivery in vivo or as genes introduced into T cells in an adoptive T cell setting.
SUMMARY OF THE INVENTION
The present invention relates to a wild-type T cell receptor against the
WT1 n, and in vitro engineered T cell receptors (TCR) that bind to the WT1
antigen with improved affinity. More ically, the present disclosure relates to the
sequences of the wild—type T cell receptor and those stabilizing and affinity mutations
selected h the display of libraries on the surface of yeast, phage, or
mammalian cells; to TCR proteins selected from these libraries for binding to an
antigen with increased affinity; and to the use of in vitro selected TCR derivatives for
therapeutic, diagnostic, or g applications.
One aspect of the invention relates to a T cell receptor, or antigen-binding
nt thereof, comprising a Va and a VB derived from a T cell clone, wherein the
T cell receptor binds to a complex of the peptide WT1 and the HLA—A2 molecule, and
wherein the T cell receptor comprises the VB amino acid ce set forth in SEQ
ID NO:1 and the Va amino acid sequence set forth in SEQ ID NO:2. In one
embodiment, a host cell that expresses the T cell receptor. In a further embodiment,
the host cell is a human T cell.
One aspect of the invention elates to a modified T cell receptor, or antigen
binding fragment thereof, comprising a Va and a VB derived from a wild type T cell
receptor, wherein the V0, the VB, or both, se a mutation in one or more
complementarity determining regions (CDRs) relative to the wild type T cell or,
wherein the modified T cell receptor binds to the peptide/MHC antigen known as
WT1/HLA-A2 (the WT1 peptide RMFPNAPYL (SEQ ID NO:6), bound to the MHC
product known as HLA-A2).
In one embodiment, the T cell receptor comprises a VB comprising an
amino acid sequence having at least 80% identity to the VB amino acid sequence set
forth in SEQ ID NO:1, wherein the T cell receptor mediates activity by binding to
WT1/H LA-A2.
In another embodiment, the modified T cell receptor comprises a ed
VB comprising an amino acid sequence having at least 80% identity to the VB amino
acid sequence set forth in SEQ ID NO:3, wherein the modified T cell receptor binds
to WT1/HLA-A2 with an affinity (KA value) of 106 M higher.
In another embodiment, the T cell receptor comprises a Va comprising an
amino acid sequence having at least 80% identity to the Va amino acid sequence set
forth in SEQ ID NO:2, wherein the T cell receptor es activity by binding to
A-A2.
In another embodiment, the modified T cell receptor comprises a modified
Va comprising an amino acid sequence having at least 80% ty to the Va amino
acid sequence set forth in SEQ ID NO:4, n the modified T cell receptor binds
to WT1/HLA-A2 with an affinity (KA value) of 106 M higher.
In one embodiment, the T cell receptor is a single-chain T cell receptor
sing the amino acid sequence set forth in SEQ ID NO:5.
In one embodiment, the T cell or ns at least one of the
mutations in CDR3B selected from 895T, 897N, l103Y, N104L of the amino acid
sequence set forth in SEQ ID NO:3.
In another embodiment, the T cell receptor contains at least one of the
mutations in CDR1a selected from V29D, SBOL, and Q31 G of the amino acid
sequence set forth in SEQ ID NO:4.
In one embodiment, the modified T cell receptor is generated by in vitro
selection of a yeast display library of mutant T cell receptors.
In r embodiment, the modified T cell receptor is expressed as a
soluble protein that binds to its target antigen.
In another embodiment, the wild-type or the modified T cell ors are
expressed in T cells for adoptive T cell therapies.
One aspect of the invention provides a therapeutic agent that targets
cancer cells that express the WT1 antigen, wherein the therapeutic agent comprises
a modified T cell receptor described herein. In one embodiment, a eutic agent
that targets cancer cells that express the WT1 antigen, wherein the therapeutic
agent ses a human T cell that expresses a modified T cell receptor described
herein. One ment provides a method of treating a subject having a cancer
that expresses the WT1 antigen comprising administering a therapeutic agent
described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the aligned amino acid sequences of various WT1/HLA—A2
ic T cell receptors from the present study. WT1 P22 represents the wild-type
sequence ed from a human T cell clone that was stimulated by WT1/HLA-A2.
The other TCR sequences were ed by yeast display methods for greater
surface levels and for higher affinities. The amino acid residues within the CDR3[3
and the CDR10i that are highlighted within rectangles were mutations in TCRs
isolated for higher affinity by yeast display. The sequences shown for the VB chain
correspond to SEQ ID NOs:1, 21, 21, 3, and 3, from top to bottom. The linker
sequence depicted is SEQ ID N028. The sequences shown for the V0. chain
pond to SEQ ID NOs:2, 22, 4, 2, and 4, from top to bottom.
Figure 2A is a nsional diagram that shows a side view of the
TCR:pepMHC complex (A6; PDB:1AO7). The variable (V) and constant (C) regions
of the d-chain and B-chain are indicated. The structure shown does not include the
Ca region of the TCR. HLA-A2 (d1, d2, (13, and 62m) is shown in gray, and the Tax
peptide (LLFGYPVYV, SEQ ID NO:7) is shown in black. The A6 TCR and the WT1
TCRs examined in the present ion all use the V02 segment (also referred to as
TRAV12 based on IMGT nomenclature).
Figure 2B is a 3-dimensional m that shows the top down view of the
TCR (CDR) footprint over the peptide-MHC LA-A2). Although no crystal
structures have been described for the WT1 TCR used in the present disclosure, this
diagonal docking orientation, with the V0 region oned over the (12 MHC helix
and the N—terminal end of the peptide, and the V6 region positioned over the G1
MHC helix and C-terminal end of the peptide, has been observed in virtually all
complexes to date.
Figure 3 is a diagram that shows a method for engineering single chain
TCRs for improved affinity against a peptide:HLA.A2. The general process used to
engineer high affinity TCRs is shown.
Figure 4 is a schematic of the yeast-display system for engineering single-
chain T cell receptor fragments (Va-linker VB or VB-linker—Va).
Figures 5A and SB show the flow cytometry histograms of the WT1 single-
chain TCR error prone library after sorting with two antibodies which recognize
conformation epitopes on V63. The WT1 error prone library was sorted sequentially
with a 1:10 on of both Therma th3.1 FITC lgG and BC th3.2 FITC lgM
antibodies, luor 647 Goat anti-mouse lgG and Goat anti-mouse lgM APC, for
a total of 3 sorts. Aliquots of yeast cells after each sort were then incubated with a
1:10 dilution of Therma th3.1 FITC lgG, luor 647 Goat anti—mouse lgG.
Gray indicates yeast cells stained with secondary antibody only (Figure 5A). The
stable clone WT1 D13, isolated after the 3rd sort, is d with a 1:10 dilution of
Therma th3.1 FITC lgG, AlexaFluor 647 Goat anti-mouse lgG (Figure 5B).
Figures 6A and 6B show the flow cytometry rams of the WT1 -
chain TCR D13 CDR1a library after sorting with WT1:HLA.A2. The WT1 D13 CDR1or
library was sorted sequentially with 100-200 nM WT1:HLA-A2 dimer (DimerX;
obtained from BD Pharmingen), APO-conjugated goat anti-mouse secondary
antibody, for a total of five sorts. Aliquots of yeast cells after each sort were then
incubated with 100 nM WT1:HLA-A2 dimer (DimerX; obtained from BD Pharmingen),
followed by njugated goat anti-mouse secondary antibody. Gray indicates
yeast cells stained with secondary antibody only (Figure 6A). Clone WT1 D13.1,
isolated after the 5th sort, is stained with 100 nM WT1:HLA-A2 dimer (DimerX;
obtained from BD Pharmingen), followed by APO-conjugated goat ouse
secondary antibody (Figure 6B).
s 7A and 7B show the flow cytometry rams of the WT1 single-
chain TCR D13.1 combined CDR3 library after g with WT1:HLA.A2. The WT1
D13.1 combined CDR3 library was sorted sequentially with 10-100 nM WT1:HLA—A2
dimer (DimerX; obtained from BD Pharmingen), APC-conjugated goat anti-mouse
secondary antibody, for a total of three sorts. Aliquots of yeast cells after each sort
were then incubated with 100 nM WT1:HLA-A2 dimer (DimerX; obtained from BD
Pharmingen), followed by APO—conjugated goat anti-mouse secondary antibody.
Gray indicates yeast cells stained with secondary antibody only (Figure 7A). The
improved binding clone WT1 D13.1.1, isolated after the 3rd sort, is stained with 100
nM WT1:HLA-A2 dimer (DimerX; obtained from BD Pharmingen), followed by APC-
conjugated goat anti-mouse secondary antibody (Figure 7B).
Figures 8A-8D show the binding properties of a high-affinity TCR, WT1
1, for WT1:HLA-A2 dimers and monomers. Figure 8A shows the flow
cytometry histograms of the high affinity scTCR WT1 D13.1.1 stained with various
concentrations of WT1 :HLA-A2 lg dimer, followed by fluorescent labeled g
antibody as a secondary. Figure 8B shows the flow try histograms of the high
affinity scTCR WT1 D13.1.1 d with various concentrations of biotinylated
WT1:HLA-A2 monomer, followed by SA-PE (1:100) secondary. Figure 8C is a line
graph that shows mean fluorescence intensity (MFI) values of histograms in Figure
7A plotted versus WT1:HLA-A2 dimer concentration. Figure 8D is a line graph that
shows mean fluorescence ity (MFI) values of histograms in Figure 8B plotted
versus WT1:HLA-A2 monomer tration.
Figures 9A-E show the binding of e high-affinity TCR WT1 D13.1.1
that was sed in E. coli. Figures 9A-D are a series of histograms that show flow
cytometry analysis of human T2 (HLA—A2+) cells incubated first with no peptide
e 9A), negative control peptide Tax (Figure QB), negative control peptide
MART-1 (Figure 9C), or peptide WT1 (Figure 9D), followed by incubation with biotin-
labeled WT1-D13.1.1 TCR. Figure SE is a graph that depicts the titration showing
that the WT1-D13.1.1 TCR had a minimum affinity (KD value) of at least 260 nM (as
washing of cells in flow cytometry results in an underestimate of affinities in this
range).
Figures 10A and 108 show the activity of wild—type P22, D13.1, D13.0.1
and D13.1.1 TCRs in mouse T cells. Isolated mouse CD8 (Figure 10A) and CD4
(Figure 108) T cells were transduced with P22, D13.1, D13.0.1 and D13.1.1 TCRs
(modified TCRs did not contain the D13 “stabilizing” mutations in the VB region:
F488 and D51G). Transduced T cells were then incubated with HLA-A2+ APCs and
various concentrations of the WT1 peptide. Following 24 hours tion, IFN-y
concentrations were measured using a standard ELISA.
Figure 11 shows that the high—affinity TCR WT1 D13.1.1 in CD8 T cells did
not show activity against a panel of WT1 structurally similar human es. WT1
structurally similar peptides were determined through searches of the human
proteome for peptides with conservative mutations at each of the 9 residues of the
WT1 peptide (SEQ ID NO:6). The 10 peptides that were ted to bind to HLA-A2
with highest affinity were then synthesized. Peptide numbers 1-10 correspond to
SEQ ID NOs:25-34, respectively. Mouse CD8 T cells were isolated and transduced
with the D13.1.1 TCR (did not contain D13 stabilizing mutations in VB , F488
and D51 G) and incubated for 24 hours with respective peptides and HLA-A2+ APCs.
Figures 12A and 12B are diagrams that illustrates exemplary therapeutic
applications of the WT1-specific TCRs. Figure 12A depicts five examples of TCR
s for use as soluble therapeutic ts: 1) -chain TCR in either a Vor—
VB orientation or VB-Va orientation (mutated high-affinity V domains are shown with
an asterisk); 2) -chain TCR fused in frame with the constant region domains of
an antibody; 3) in—frame immunoglobulin fusion to either the constant region of the
light chain or the heavy chain; 4) single-chain TCR (or the immunoglobulin fusions
shown in 2 and 3) directly coupled to a drug; and 5) single—chain TCR linked in—frame
with a single-chain Fv (VL-linker—VH) to generate a bispecific agent. Figure 128
depicts two es of cellular based therapies that would use the ffinity
variable domains (V) isolated by yeast display, or the ype TCR V domains may
also be used for adoptive T cell therapies with human T cells). The TCRs are cloned
into mammalian cell vectors, for expression by T cells in adoptive T cell therapy as:
1) single-chain receptors in chimeric antigen receptors (CAR) and 2) full length or and
[3 TCRs.
BRIEF DESCRIPTION OF THE SEQUENCES
SEQ ID NO:1 is the amino acid sequence of a wild-type V0 region of the
TCR (P22) that binds to WT1/HLA-A2.
SEQ ID NO:2 is the amino acid sequence of a wild-type Vor region of the
TCR (P22) that binds to WT1/HLA-A2.
SEQ ID NO:3 is the amino acid sequence of a modified V0 region of the
TCR (D13.1.1) that binds with high-affinity to WT1/HLA-A2.
SEQ ID NO:4 is the amino acid sequence of a ed Vor region of the
TCR (D13.1.1) that binds with high-affinity to WT1/HLA-A2.
SEQ ID NO:5 is the amino acid ce of a single-chain TCR (WT1-
D13.1.1) that binds with high-affinity to WT1/HLA-A2.
SEQ ID NO:6 is the amino acid sequence of the WT-1 antigen.
SEQ ID NO:7 is the amino acid sequence of the Tax n.
SEQ ID NO:8 is the amino acid sequence of the .
SEQ ID NO:9 is the is the polynucleotide sequence of the primer Splice
SEQ ID NO:10 is the is the polynucleotide sequence of the reverse primer
used to generate the WT1-D13 CDR10L library PreSOE #1 .
SEQ ID NO:11 is the is the polynucleotide sequence of the forward primer
used to generate the WT1-D13 CDR1a library PreSOE #2.
SEQ ID NO:12 is the is the polynucleotide sequence of the primer T7.
SEQ ID NO:13 is the is the polynucleotide sequence of the reverse primer
used to generate the PreSOE #1 of the WT1-D131 CDR3 [51 library
SEQ ID NO:14 is the is the polynucleotide sequence of the forward primer
used to generate the PreSOE #2 of the WT1-D131 CDR3 B1 library
SEQ ID NO:15 is the the polynucleotide ce of the reverse primer
used to generate the PreSOE #1 of the WT1-D13.1 CDR3 [32 library.
SEQ ID NO:16 is the is the polynucleotide ce of the fon/vard primer
used to generate the PreSOE #2 of the WT1-D13.1 CDR3 [32 library.
SEQ ID NO:17 is the the polynucleotide sequence of the reverse primer
used to generate the PreSOE #1 of the 3.1 CDR3 of y
SEQ ID NO:18 is the is the polynucleotide sequence of the forward primer
used to generate the PreSOE #2 of the WT1-D13.1 CDR3 of library
SEQ ID NO:19 is the is the polynucleotide sequence of the reverse primer
used to generate the PreSOE #1 of the WT1-D13.1 CDR3 02 library
SEQ ID NO:20 is the is the polynucleotide sequence of the fon/vard primer
used to generate the PreSOE #2 of the WT1-D13.1 CDR3 a2 library.
SEQ ID NO:21 is the amino acid sequence of a modified V0 region of the
TCR (D13) that binds with high-affinity to WT1/HLA-A2.
SEQ ID NO:22 is the amino acid sequence of a modified Vor region of the
TCR (D13) that binds with high-affinity to WT1/HLA-A2.
SEQ ID NO:23 is the amino acid sequence of an influenza A peptide.
SEQ ID NO:24 is the amino acid sequence of a variant influenza A
peptide.
SEQ ID NOs:25-34 are the amino acid sequences often WT1 variant
peptides.
DETAILED DESCRIPTION
The ing description is intended to facilitate understanding of the
disclosure but is not intended to be limiting.
In general, the terms and phrases used herein have their art-recognized
meaning, which can be found by reference to standard texts, journal references and
contexts known to those skilled in the art. The following definitions are provided to
clarify their ic use in the context of the disclosure.
As used herein, “linked” refers to an association n two groups,
which can be a covalent or non—covalent association. Groups may be linked using a
variable length peptide chain, a non-amino acid chemical group or other means as
known in the art. A linker region can be an amino acid ce that operably links
two functional or structural domains of a protein or peptide.
As used herein, the term “chemotherapeutic agent” refers to any
nce capable of reducing or ting the growth, proliferation, or spread of a
cancer cell, a population of cancer cells, tumor, or other malignant tissue. The term
is intended also to encompass any antitumor or anticancer agent.
As used herein, the term “effective amount” is intended to encompass
contexts such as a pharmaceutically effective amount or eutically effective
amount. For example, in certain embodiments, the effective amount is capable of
achieving a beneficial state, beneficial outcome, functional activity in a screening
assay, or improvement of a al condition.
As used herein, the term “cancer cell” is intended to encompass definitions
as broadly understood in the art. In one embodiment, the term refers to an
abnormally regulated cell that can contribute to a clinical condition of cancer in a
human or animal. In one embodiment, the term can refer to a cultured cell line or a
cell within or derived from a human or animal body. A cancer cell can be of a wide
variety of differentiated cell, tissue, or organ types as is understood in the art.
Particular examples of cancer cells include breast cancer, colon cancer, skin cancer,
ovarian cancer, leukemia, lung cancer, liver cancer, testicular cancer, esophageal
cancer, and other types of cancer.
As used herein, “treating” or “treatment” refers to an ch for
obtaining beneficial or d results, ing and preferably clinical results.
Treatment can refer to either the amelioration of ms of the disease or
condition, or the ng of the ssion of the e or ion.
As used herein, "prevention" or “preventing” refers to an approach for
preventing, inhibiting, or reducing the likelihood of, the onset or recurrence of a
disease or condition. It also refers to preventing, inhibiting, or reducing the likelihood
of, the occurrence or recurrence of the ms of a disease or condition, and it
also includes reducing the intensity, effect, symptoms and/or burden of a disease or
condition prior to onset or recurrence of the disease or condition.
As used herein, “inhibiting cell growth” or “inhibiting proliferation of cells”
refers to reducing or halting the growth rate of cells. For example, by inhibiting the
growth of tumor cells, the rate of increase in size of the tumor may slow. In other
embodiments, the tumor may stay the same size or se in size, i.e., regress.
In particular embodiments, the rate of cell growth or cell proliferation is inhibited by at
least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at
least 80%, or at least 90%.
The terms “wild type” and “wt” are used interchangeably herein and are
used in reference to a TCR having an amino acid sequence or a polynucleotide
encoding the variable regions ed from a naturally occurring or non-modified
TCR, e.g., the original or parent T cell clone, with specificity for the antigen.
In the figures and tables that present amino acid sequences, the wild type
is sometimes designated “wt”. In the sequences presented below the top sequence,
a dash indicates the amino acid is the same as that present in the wt or top
sequence of the alignment. A letter indicates a substitution has been made in that
position from the top sequence.
As used herein, the terms “modified”, “variant, , mutated” and
“derived” T cell receptor refer to TCR ces of the le regions having one
or more mutations compared to the original or wild type T cell clone. Examples of
modified TCRs include higher affinity TCRs.
A coding sequence is the part of a gene or cDNA which codes for the
amino acid sequence of a protein, or for a functional RNA such as a tRNA or rRNA.
Complement or complementary ce means a sequence of
tides that forms a hydrogen-bonded duplex with another sequence of
nucleotides according to Watson-Crick base-pairing rules.
Downstream refers to a relative position in DNA or RNA and is the region
toward the 3' end of a strand.
Expression refers to the ription of a gene into structural RNA (rRNA,
tRNA) or messenger RNA (mRNA) and subsequent translation of an mRNA into a
protein.
Two nucleic acid sequences are heterologous to one another if the
ces are derived from separate organisms, whether or not such organisms are
of different species, as long as the sequences do not naturally occur together in the
same arrangement in the same organism.
Homology refers to the extent of identity between two nucleotide or amino
acid sequences.
An amino acid sequence that is onally equivalent to a specifically
exemplified TCR ce is an amino acid sequence that has been modified by
single or multiple amino acid substitutions, by addition and/or deletion of amino
acids, or where one or more amino acids have been chemically modified, but which
nevertheless retains the binding specificity and high affinity binding activity of a cell
bound or a soluble TCR protein of the t disclosure. Functionally equivalent
nucleotide sequences are those that encode polypeptides having substantially the
same biological activity as a specifically exemplified cell—bound or soluble TCR
n. In the context of the present disclosure, a soluble TCR protein lacks the
portions of a native cell—bound TCR and is stable in solution (i.e., it does not
generally aggregate in solution when d as described herein and under
standard ions for protein solutions).
The term “isolated” refers to a composition, compound, substance, or
molecule altered by the hand of man from the natural state. For e, a
composition or substance that occurs in nature is isolated if it has been changed or
removed from its original environment, or both. For example, a polynucleotide or a
polypeptide naturally t in a living animal is not isolated, but the same
polynucleotide or polypeptide separated from the coexisting materials of its natural
state is isolated, as the term is employed herein.
A nucleic acid construct is a nucleic acid molecule which is isolated from a
naturally occurring gene or which has been modified to contain segments of nucleic
acid which are combined and juxtaposed in a manner which would not otherwise
exist in nature.
Nucleic acid molecule means a single— or double—stranded linear
polynucleotide containing either deoxyribonucleotides or ribonucleotides that are
linked by phosphodiester bonds.
Two DNA sequences are operably linked if the nature of the linkage does
not interfere with the ability of the sequences to affect their normal functions relative
to each other. For instance, a promoter region would be operably linked to a coding
sequence if the promoter were capable of effecting transcription of that coding
sequence.
A polypeptide is a linear polymer of amino acids that are linked by peptide
bonds.
The term “promoter” refers to a cis-acting DNA sequence, generally 80-
120 base pairs long and located upstream of the initiation site of a gene, to which
RNA polymerase may bind and te t transcription. There can be associated
additional transcription regulatory sequences which e on/off regulation of
transcription and/or which e (increase) expression of the downstream coding
sequence.
A recombinant nucleic acid molecule, for instance a inant DNA
molecule, is a novel nucleic acid sequence formed in vitro through the on of two
or more nonhomologous DNA molecules (for example a recombinant plasmid
containing one or more inserts of foreign DNA cloned into at least one g site).
The terms “transformation” and “transfection” refer to the directed
modification of the genome of a cell by the external application of purified
recombinant DNA from another cell of different genotype, leading to its uptake and
ation into the subject cell’s genome. ln bacteria, the inant DNA is not
typically integrated into the bacterial chromosome, but d replicates
autonomously as a d. The terms “transformed” and “transfected” are used
interchangeably herein. For example, a T cell may be transfected with a DNA
sequence encoding a modified or high affinity TCR bed herein prior to adoptive
T cell treatment.
Upstream means on the 5' side of any site in DNA or RNA.
A vector is a nucleic acid molecule that is able to replicate autonomously
in a host cell and can accept foreign DNA. A vector carries its own origin of
replication, one or more unique recognition sites for restriction endonucleases which
can be used for the insertion of foreign DNA, and usually selectable markers such as
genes coding for antibiotic resistance, and often ition sequences
(e.g.,promoter) for the expression of the inserted DNA. Common vectors include
plasmid vectors and phage vectors.
A high affinity T cell receptor (TCR) is an engineered TCR with stronger
binding to a target ligand than the wild type TCR. Some es of high affinity
include an equilibrium binding constant for a target ligand of between about 10'6 M
and 10'12 M and all individual values and ranges therein. This range encompasses
affinities between those reported to be wild type affinities (10'4 to 10'6 M), and those
which have been isolated by directed evolution (about 10'12 M).
A cytokine is a protein, e or glycoprotein made by cells that affect
other cells.
Mammal includes both human and non-human mammals.
It will be appreciated by those of skill in the art that, due to the degeneracy
of the genetic code, numerous functionally equivalent nucleotide sequences encode
the same amino acid sequence.
T Cell Receptors
The T cell receptor (TCR) is composed of two chains (a8 or y8) that pair on
the surface of the T cell to form a heterodimeric receptor. The dB TCR is expressed
on most T cells in the body and is known to be involved in the recognition of MHC-
cted antigens. The molecular genetics, structure, and biochemistry of a8 TCRs
have now been studied thoroughly. Each or and [3 chain is composed of two domains:
Constant domains (C) that anchor the protein in the cell membrane and that
associate with invariant subunits of the CD3 signaling apparatus, and Variable
domains (V) that confer antigen ition h six loops, called
complementarity determining regions (CDR). Each of the V domains has three
CDRs. These CDRs interact with a complex between an antigenic peptide bound to
a protein encoded by the major histocompatibility complex (pepMHC) (Davis and
Bjorkman (1988) Nature, 334, 2; Davis et al. (1998) Annu Rev Immunol, 16,
523-544; Murphy (2012), xix, 868 p.).
The molecular genetics of the TCR have revealed a s of genetic
recombination between multiple genes that combine to form the coding region of the
V domains. The process is analogous to antibody development in which the heavy
and light chain genes rearrange to generate the tremendous diversity exhibited by B
cell-derived antibodies (Tonegawa (1988) In Vitro Cell Dev Biol, 24, 253-65). In the
case of T cells, the or chain V domain is formed by the rearrangement of one V
region (among about 75 in humans) to one Joining (J) gene segment (among about
61 in ) e 5.8, Janeway, 8th edition). The [3 chain V domain is formed by
the rearrangement of one V region (among about 52 in humans) to one Diversity (D)
gene (among 2 in humans) to one Joining (J) gene segment (among 13 in humans)
(Figure 5.8, (Murphy (2012), xix, 868 p.)). The ons of the VorJor and VBDBJB
gene ngements encode the CDR3 loops of each chain, and they contribute to
the tremendous diversity of the (18 TCR, with a theoretical limit of over 1015 different
TCRs (Davis and Bjorkman (1988) Nature, 334, 395-402), well above the able
diversity in a human because there are only about 1011 T cells total (Mason (1998)
Immunol Today, 19, 395-404). The possible CDR1 and CDR2 diversity of each chain
is represented by the number of V genes, as these loops are encoded within the V
gene, and TCRs do not undergo somatic on in vivo. Although the diversity of
CDR1 and CDR2 loops are relatively limited compared to CDR3 loops, there have
been a number of examples shown where there has been selection for particular V
regions based on the peptide antigen and/or MHC product.
Class | MHC products bind to es of 8 to 10 amino acids in length
and they are expressed on all ted cells in the body (reviewed by (Rock and
rg (1999) Annu Rev Immunol, 17, 739-79)). s all the binding energy of
an antibody-antigen interaction is focused on the foreign antigen, a substantial
fraction of the binding energy of the TCR-peptide:MHC is directed at the self-MHC
le (Manning and Kranz (1999) Immunology Today, 20, 417—422). In fact, more
recent studies have suggested that particular residues of the CDR1 and/or CDR2
loops have evolved to interact with particular es on the MHC helices, y
providing a basal affinity for MHC, accounting for the s of MHC—restriction
(Garcia et al. (2009) Nat l, 10, 143-7; Marrack et al. (2008) Annu Rev
Immunol, 26, 171-203).
There has been interest in using TCRs that have affinities for a peptide-
MHC antigen (class I) above the normal range (so called higher affinity TCRs) in
order to: 1) drive the activity of CD4 helper T cells (which lack the CD8 ptor)
or 2) develop soluble TCRs that could be used for direct targeting of a cell, by
attaching an “effector” molecule (e.g., antibody Fc regions, a toxic drug, or an
antibody scFv such as an anti-CD3 antibody, to form a ific protein)((Ashfield
and Jakobsen (2006) lDrugs, 9, 554-9; Foote and Eisen (2000) Proc Natl Acad Sci
USA, 97, 10679-81; Holler et al. (2000) Proc Natl Acad Sci U S A, 97, 5387-92;
Molloy et al. (2005) Curr Opin Pharmacol, 5, 438—43; Richman and Kranz (2007)
Biomol Eng, 24, 361-73). This approach also could overcome a problem faced by
some cancer patients, whereby their T cells do not express TCRs with adequate
specificity and binding affinity to potential tumor antigens (in part due to the thymic
and peripheral ses of tolerance). For example, over 300 MHC-restricted, T
cell—defined tumor antigens have now been identified
(cancerimmunity.org/peptide/)(Boon and Old (1997) Curr Opin Immunol, 9, 681-3;
Cheever et al. (2009) Clin Cancer Res, 15, 5323-37). These tumor antigens include
mutated peptides, differentiation antigens, and overexpressed antigens, all of which
could serve as targets for therapies. Because the majority of the cancer antigens
described to date were derived from intracellular proteins that can only be targeted at
the cell surface in the context of an MHC molecule, TCRs make the ideal candidate
for therapeutics as they have evolved to recognize this class of antigen.
Similarly, TCRs can detect peptides derived from viral proteins that have
been naturally processed in infected cells and displayed by an MHC molecule on the
cell e. Many viral antigen targets have been identified over the past 25 years,
including peptides d from viral genomes in HIV and HTLV (e.g., Addo et al.
(2007) PLoS ONE, 2, e321; Tsomides et al. (1994) J Exp Med, 180, 3; Utz et
al. (1996) J Virol, 70, 843-51). However, patients with these diseases may lack the
optimal TCRs for binding and destruction of the infected cells. Finally, it is possible
that TCRs could be used as receptor antagonists of autoimmune targets, or as
delivery agents to immunosuppress the local immune cell response, in a process
that would be highly specific, thereby avoiding l immune suppression ((Molloy
et al. (2005) Curr Opin Pharmacol, 5, 438-43; Stone et al. (2012) Protein
Engineering)).
Modified T Cell Receptors
ed evolution has been used to generate TCRs with higher affinity for
a specific pepMHC. The three different display methods that have been used are
yeast display (Holler et al. (2003) Nat l, 4, 55-62; Holler et al. (2000) Proc
Natl Acad Sci U S A, 97, 2), phage display (Li et al. (2005) Nat Biotechnol, 23,
349-54), and T cell display in et al. (2008) J Immunol Methods, 339, 175-84).
In all three approaches, the process involves the engineering of a TCR that exhibits
the normal, low ty of the wild-type TCR, so mutants of the TCR had increased
affinity for the specific pepMHC (i.e., for the original antigen that the T cells were
specific for). Thus, the wild-type TCR was used as a template for producing
mutagenized libraries in one or more of the CDRs, followed by selection of mutants
with higher affinity, by binding to the cognate peptide-MHC n. It is well known
in the art that such in vitro, directed evolution, is necessary in order to engineer
affinities that are more than just a few fold above the wild type affinity.
] Yeast display allows for the protein of interest to be expressed on the
surface as an Aga2-fusion (Boder and Wittrup (1997) Nat. Biotech., 15, 553-557;
Boder and Wittrup (2000) s Enzymol, 328, 430-44). This system has been
used successfully in the engineering of higher affinity TCRs, -chain antibodies,
fibronectin, and other proteins. In the yeast y system, the TCR has been
yed as a stabilized single-chain protein, in VB-linker-Va or Va-linker-VB forms
(Aggen et al. (2011) Protein Engineering, Design, & Selection, 24, 361-72; Holler et
al. (2000) Proc Natl Acad Sci U S A, 97, 5387-92; Kieke et al. (1999) Proc Natl Acad
Sci U S A, 96, 5651-6; Richman et al. (2009) Mol l, 46, 902-16; Weber et al.
(2005) Proc Natl Acad Sci U S A, 102, 19033-8), or as a two-chain heterodimer
(Aggen et al. (2011) Protein Engineering, Design, & Selection, 24, 361—72; Richman
et al. (2009) Mol Immunol, 46, 902-16). Two mouse TCRs have been engineered for
higher affinity using this system: 2C (MHC class-l restricted) and 3.L2 (MHC class-ll
restricted) (Holler et al. (2000) Proc Natl Acad Sci U S A, 97, 5387—92; Weber et al.
(2005) Proc Natl Acad Sci U S A, 102, 19033-8). Human TCR single-chain VorVB
nts (called sch or scTCR) have also recently been developed by taking
advantage of the exceptional stability of the human Vor region called Va2, also
known as TCRA12 by IMGT nomenclature (Aggen et al. (2011) Protein Engineering,
Design, & Selection, 24, 361-72). In this case, in vitro engineered, high-affinity T cell
receptors in a single-chain format were used to isolate human stabilized sch
fragments (VB-linker-Va), which could be expressed as stable proteins, both on the
surface of yeast and in soluble form from E. coli. The TCRs ed two ized,
human sch nts, the A6 sch that is specific for a peptide derived from the
human T cell trophic virus Tax protein and the 868 sch that is specific for a
peptide derived from the human immunodeficiency virus Gag protein (peptide:
SL977-85). Both of these TCRs used the V02 gene (IMGT: TRAV12 family), but they
had CDR30i, CDR1B, CDR2[3, and CDR3[3 residues derived from the original T cell
clone from which the TCRs were isolated. Thus, the higher affinity mutants of these
scTCRs were each derived from their original (parental) TCR against their cognate
peptide-MHC antigens.
In a second , phage display, the protein of interest is fused to the
N-terminus of a viral coat protein (Scott and Smith (1990) Science, 249, 386-90).
Various TCRs, including those called A6, 868, and 1G4 (MHC class-l cted),
have been engineered for higher affinity using this method (Li et al. (2005) Nat
Biotechnol, 23, 349—54; Sami et al. (2007) n Eng Des Sel, 20, 397-403; Varela-
Rohena et al. (2008) Nat Med, 14, 1390-5). Phage display of these TCRs was
enabled by introduction of a non-native disulfide bond between the two C domains in
order to promote pairing of the or and B chains. This system thus uses full-length
(VuCaNBCB) heterodimeric proteins derived from the al T cell clones for
engineering against their cognate peptide-MHC.
A third system that has been ed for the engineering of TCRs is
mammalian cell display (Chervin et al. (2008) J l Methods, 339, 175-84;
Kessels et al. (2000) Proc Natl Acad Sci U S A, 97, 14578—83). This system uses a
retroviral vector to introduce the TCR d and B-chains into a TCR-negative T cell
hybridoma. In one study (Kessels et al. (2000) Proc Natl Acad Sci U S A, 97, 14578-
83), the selected mutant TCR was shown to bind to a peptide that was structurally
very similar to the cognate peptide (ASNENMDAM versus ASNENMETM; SEQ ID
NOs:23 and 24, respectively). In the other study, the affinity of the mutant TCR was
shown to be increased for the e pepMHC (Chervin et al. (2008) J Immunol
Methods, 339, 175-84). It has been shown in many studies that such higher affinity
TCRs also exhibit higher affinities against urally similar variants of the cognate
peptide (e.g.,(HolIer et al. (2003) Nat Immunol, 4, 55-62)). In the mammalian cell
y system, introduced TCRs were expressed on the surface in its native
conformation, in complex with CD3 subunits, ng for a fully functional T cell
(signaling competent). Full-length, dimeric TCRs in their native host were thus
engineered using this method.
High—Affinity TCRs that Bind to WT1/HLA-A2
The present invention provides for a wild type TCR and various highaffinity
TCRs against the well-known cancer antigen WT1/HLA-A2. In certain
embodiments, the ered TCRs can be used in soluble form for targeted
delivery in vivo, or as recombinantly expressed by T cells in an adoptive er
method or treatment. In a ular embodiment, a single-chain VaVB form of the
TCR ) scaffold can be prepared and used with a payload such as a cytokine,
toxin, radioisotope, chemotherapeutic agent, or drug (similar to antibody—drug
conjugates) to deliver the effector molecule to the location where the TCR binds
(e.g., tumor). The TCR can also be used in cell therapies, such as adoptive transfer
of CD4+ T cells, CD8+ T cells, and/or natural killer (NK) cells, to mediate a response
against cancer cells that express WT1. The scTCR scaffolds provided herein can
also be used for sis of, e.g., malignant or viral-infected cells through
identification of, e.g., neoplastic or viral-associated urface antigens by covalent
linkage, for example through amine-reactive or dryl-reactive amino acid side
chains of the TCR, to a detectable group, such as a radioisotope or fluorescent
moiety.
In one embodiment, the scTCR proteins described herein are yable
on the surface of yeast, phage, or mammalian cells and can be used to engineer
TCRs with even higher affinity to the WT1 antigen. In one embodiment, the scTCR
ns bed herein can be expressed in a prokaryotic cell, such as
Escherichia coli, Aspergil/us niger, Aspergi/lus ficuum, Aspergi/lus awamori,
Aspergillus Oiyzae, Trichoderma reesei, Mucor miehei, Kluyveromyces lactis, Pichia
pastoris, romyces cerevisiae, Bacillus subti/is or Bacillus lichen/formis, insect
cells (e.g., hila melanogaster), mammalian cells including cell lines such as
Chinese hamster ovary cell lines (CHO), or plant species (e.g., canola, soybean,
corn, , barley, rye, wheat) for example, or other art-known protein expression
sources and produced in large quantities. The TCR can also be used, for e
and by way of example only, to detect the specific peptide/MHC on the surface of a
cell. In one embodiment, the scTCR genes disclosed can be linked by use of suitable
peptide sequences, encoded within the DNA construct, to the genes for signaling
domains and introduced into T cells that can eliminate the targeted cells. These
constructs have been termed ic antigen receptors (CARs), which are now
widely used in the field, including the use of CARs that contain a scTCR.
In the single-chain VaVB TCR proteins provided, the variable alpha and
variable beta chains are connected using any suitable peptide linker, including those
known in the art such as with antibody single—chain Fv linkages (Bird et al. (1988)
Science, 242, 423-426; Holliger et al. (1993) Proc Natl Acad Sci U S A, 90, 6444-8;
Hoogenboom (2005) Nat Biotechnol, 23, 1105-16; Turner et al. (1997) J Immunol
Methods, 205, 43-54). In one embodiment, a soluble human single-chain TCR
having the structure: Vd-L-VB or d, wherein L is a linker peptide that links V8
with Va, VB is a TCR variable [3 region, and Va is a TCR variable (1 region is
provided.
In one embodiment, the VBVa TCR is called WT1 D13.1.1 where VB is a
TCR variable [3 region of group 3, and Vd2 is a TCR le or region of group 2
(Utz, U., et al., 1996)(Aggen, D.A., et al., 2011).
In one embodiment, the linker peptide contains more than 5 lysine
residues. In one embodiment, the linker peptide contains between 5 and 30 amino
acids. In one embodiment, the linker peptide has an amino acid sequence of
GSADDAKKDAAKKDGKS (SEQ ID NO:8). In one embodiment, the sc VBVOL TCR
provided does not contain a constant region. When the terminology sc VBVQ TCR is
used herein, it is understood that sc VBVo TCR is also ed as the terminology is
understood and used in the art. Thus, the VB and Va chains can be connected to
each other in any configuration through the .
In an aspect of the disclosure, the VBVOL TCR of the disclosure binds
specifically to a ligand with an equilibrium binding constant KB of between about 10'6
M and 10'12 M. In one embodiment of this aspect of the disclosure, the ligand is a
peptide/MHC ligand. In one embodiment, the VBVa TCR of the disclosure has
enhanced affinity toward a ligand compared to the affinities of normal, wild type
TCRs.
ically Active Groups
] Also provided are VBVa TCR proteins as described herein which includes
a biologically active group. As used herein, “biologically active group” is a group that
causes a measurable or detectable effect in a biological . In one embodiment,
the biologically active group is selected from: an anti-tumor agent such as, but not
limited to, angiogenesis inhibitors, enzyme inhibitors, ubule inhibitors, DNA
intercalators or cross-linkers, DNA synthesis inhibitors; a ne such as, but not
limited to |L-2, lL-15, , |L-12, TNF-0i, IFN-y or LT-0i (Schrama et al. (2006)
Nat Rev Drug Discov, 5, ; Wong et al. (2011) Protein Eng Des Sel, 24, 373-
83); an anti-inflammatory group such as, but not limited to, TGF-B, |L-37, |L-10 (Nold
et al. (2010) Nat Immunol, 11, 1014-22; Stone et al. (2012) Protein Engineering), a
radioisotope such as, but not limited to, 90v or 131i (Reichert and Valge—Archer (2007)
Nat Rev Drug Discov, 6, 349-56); a toxin such as, but not limited to, Pseudomonas
exotoxin A, diphtheria toxin, or the A chain of ricin (Pastan et al. (2006) Nat Rev
Cancer, 6, 559-65; Schrama et al. (2006) Nat Rev Drug Discov, 5, ); a drug,
or an antibody such as a single-chain Fv.
In one embodiment of this aspect of the disclosure, the biologically active
group is a cytotoxic molecule, mes referred to as a drug (e.g., in the term
“antibody drug conjugate”). As used herein, oxic” means toxic to cells.
Examples of cytotoxic les include, but are not limited to, doxorubicin,
methotrexate, mitomycin, 5-fluorouracil, duocarmycin, atins, maytansines,
calicheamicins and analogs of the above molecules (Jarvis (2012) Chemical and
Engineering News, 90, 12-18; Litvak-Greenfeld and Benhar (2012) Adv Drug Deliv
Rev; Ricart and Tolcher (2007) Nat Clin Pract Oncol, 4, 245-55). Cytotoxic molecules
do not need to cause complete cell death, but rather, a able or detectable
inhibition of growth or decrease in cell activity.
In one embodiment, a TCR described herein is linked to an enzyme
capable of converting a prodrug into a drug. This is useful, for example, by allowing
the active form of the drug to be created at the location ed by the TCR (e.g., at
the site of a tumor).
In one embodiment, the biologically active group is bound to the single—
chain TCR through a linker, which may be accomplished through standard chemical
reactions such as with free amine groups or dryl groups of the TCR.
In another embodiment, the TCR is attached to a single-chain antibody
fragment (scFv) to generate a ific agent. Bispecific antibodies that contain one
scFv against a tumor antigen, and one against the CD3 molecule of the T cell have
now been used successfully in the clinic (Bargou et al. (2008) Science, 321, 974-7).
In addition, a bispecific agent ning a TCR and a scFv against CD3 has also
been reported (Liddy et al. (2012) Nat Med, 18, 980-7).
Also provided is a single-chain VBVoc TCR as described herein which
includes a detectable group. In one embodiment, the detectable group is one that
can be detected by spectroscopic or enzyme-based methods. In one embodiment,
the detectable group is a scent group, such as, but not limited to fluorescein, R-
phycoerythrin (PE), PE-Cy5, PE-Cy7, Texas red, or allophycocyanin (APC); a
radiolabeled group such as, but not d to, 125i, 32P, 99mTc; an absorbing
group, or an enzyme with ties that generate detectable products such as, but
not limited to, horseradish dase, or alkaline phosphatase.
As known in the art, a biologically active group, detectable group or other
group ed to the TCR can be attached using a flexible peptide linker or by
chemical conjugation, and can be covalently or noncovalently attached to the TCR.
] Also provided herein is a human TCR for use in a method of treating or
preventing cancer in a mammal, comprising administering an effective amount of a
modified TCR linked to a eutically effective molecule to a mammal. In a
particular embodiment, the mammal is human. In another embodiment, the mammal
is a companion animal (e.g., a dog, cat, , rodent, horse) or a livestock animal
(e.g., a cow, horse, pig).
Also provided is an isolated single-chain TCR (scTCR) as described
herein, and a method for producing the single-chain TCR in E. coli. Also provided is
a pharmaceutical composition comprising a scTCR as described herein and a
pharmaceutically acceptable r.
Also provided is the sc VaVB TCRs described herein which have been
linked to ing domains that yields an active TCR on the surface of a T cell. In
one embodiment, this scTCR can be used in a method of treating cancer in a
mammal, comprising: cloning the TCR into a vector, introducing the vector into T
cells of a patient, and adoptive erring of the T cells back into a patient.
ed TCR Polypeptides and Polynucleotides
The disclosure contemplates a DNA vector that includes at least one DNA
segment encoding a single—chain T cell receptor (scTCR).
Those of skill in the art, through standard mutagenesis techniques,
conjunction with the assays described herein, can obtain altered TCR sequences
and test them for particular binding ty andlor specificity. Useful mutagenesis
techniques known in the art include, without limitation, de novo gene synthesis,
oligonucleotide-directed mutagenesis, -specific nesis, linker-scanning
mutagenesis, and site-directed mutagenesis by PCR (see e.g., Sambrook et al.
(1989) and Ausubel et al. (1999)).
In obtaining modified TCR coding sequences, those of ordinary skill in the
art will recognize that rived proteins may be modified by certain amino acid
substitutions, additions, deletions, and post-translational modifications, without loss
or reduction of ical activity. In particular, it is well known that conservative
amino acid tutions, that is, substitution of one amino acid for another amino
acid of similar size, charge, polarity and conformation, are unlikely to significantly
alter protein function. The 20 standard amino acids that are the constituents of
ns can be broadly categorized into four groups of conservative amino acids as
follows: the nonpolar (hydrophobic) group includes e, isoleucine, leucine,
methionine, phenylalanine, proline, tryptophan and valine; the polar rged,
neutral) group includes asparagine, cysteine, glutamine, glycine, serine, threonine
and tyrosine; the positively charged (basic) group contains arginine, ine and
lysine; and the negatively charged (acidic) group contains aspartic acid and glutamic
acid. Substitution in a protein of one amino acid for r within the same group is
unlikely to have an adverse effect on the biological ty of the n.
] In one embodiment, a scTCR of the disclosure may contain onal
mutations in any region or regions of the variable domain that results in a stabilized
protein. In one embodiment, one or more additional ons is in one or more of
CDR1, CDR2, HV4, CDR3, FR2, and FR3. The regions used for mutagenesis can be
determined by directed evolution, where crystal structures or molecular models are
used to generate regions of the TCR which interact with the ligand of interest
(antigen, for example). In other examples, the variable region can be reshaped, by
adding or deleting amino acids to engineer a desired interaction between the scTCR
and the ligand.
] Polypeptides of the invention include modified TCRs, and antigen binding
fragments thereof (e.g., scTCR), and chimeric antigen receptors (CARS). The terms
"polypeptide protein" and "peptide" and "glycoprotein" are used interchangeably
and mean a polymer of amino acids not limited to any particular length. The term
does not exclude modifications such as myristylation, sulfation, glycosylation,
phosphorylation and addition or deletion of signal sequences. The terms
"polypeptide" or "protein" means one or more chains of amino acids, wherein each
chain comprises amino acids ntly linked by peptide bonds, and wherein said
ptide or protein can comprise a ity of chains non-covalently and/or
covalently linked together by peptide bonds, having the sequence of native proteins,
that is, proteins produced by naturally-occurring and specifically non-recombinant
cells, or genetically-engineered or recombinant cells, and comprise molecules having
the amino acid sequence of the native protein, or molecules having deletions from,
additions to, and/or substitutions of one or more amino acids of the native sequence.
The terms "polypeptide" and "protein" specifically encompass the modified TCRs, or
antigen-binding fragments thereof, of the present disclosure, or sequences that have
deletions from, additions to, and/or substitutions of one or more amino acid of a
modified TCR, or antigen g fragment thereof. Thus, a "polypeptide" or a
"protein" can comprise one (termed "a r") or a plurality (termed “a multimer")
of amino acid chains.
The term “isolated n" referred to herein means that a subject protein
(1) is free of at least some other proteins with which it would typically be found in
nature, (2) is essentially free of other proteins from the same source, e.g., from the
same species, (3) is expressed by a cell from a ent species, (4) has been
ted from at least about 50 percent of polynucleotides, lipids, carbohydrates, or
other materials with which it is associated in nature, (5) is not associated (by
covalent or noncovalent interaction) with portions of a protein with which the "isolated
protein" is associated in nature, (6) is operably associated (by covalent or
noncovalent interaction) with a polypeptide with which it is not associated in nature,
or (7) does not occur in nature. Such an isolated protein can be encoded by genomic
DNA, cDNA, mRNA or other RNA, of may be of synthetic origin, or any combination
thereof. In certain ments, the isolated protein is substantially free from
proteins or ptides or other contaminants that are found in its natural
environment that would interfere with its use (therapeutic, diagnostic, prophylactic,
research or otherwise).
In particular embodiments, a subject modified TCR may have: a) a TCR
alpha chain variable region having an amino acid sequence that is at least 80%
identical, at least 85% identical, at least 90%, at least 95% or at least 98% or 99%
identical, to the alpha chain le region of a modified TCR described herein; and
b) a beta chain variable region having an amino acid sequence that is at least 80%
identical, at least 85%, at least 90%, at least 95% or at least 98% or 99% identical, to
the beta chain variable region of a modified TCR described herein.
In particular embodiments, the modified TCR may comprise: a) a TCR
alpha chain variable region comprising: i. a CDR1 region that is identical in amino
acid sequence to the alpha chain CDR1 region of a selected TCR described herein;
ii. a CDR2 region that is identical in amino acid sequence to the alpha chain CDR2
region of the selected TCR; and iii. a CDR3 region that is identical in amino acid
sequence to the alpha chain CDR3 region of the ed TCR; and b) a beta chain
variable region sing: i. a CDR1 region that is identical in amino acid sequence
to the beta chain CDR1 region of the selected TCR; ii. a CDR2 region that is
identical in amino acid ce to the beta chain CDR2 region of the selected TCR;
and iii. a CDR3 region that is identical in amino acid sequence to the beta chain
CDR3 region of the selected TCR; wherein the TCR specifically binds a the WT1
antigen. In a further embodiment, the modified TCR, or antigen-binding fragment
thereof, is a variant modified TCR wherein the variant comprises an alpha chain and
a beta chain cal to the ed modified TCR except for up to 8, 9, 10, 11, 12,
13, 14, 15, or more amino acid tutions in the CDR regions of the V alpha and V
beta regions. In this regard, there may be 1, 2, 3, 4, 5, 6, 7, 8, or in certain
embodiments, 9, 10, 11, 12, 13, 14, 15 more amino acid substitutions in the CDR
regions of the selected variant modified TCR. Substitutions may be in CDRs either in
the V alpha and/or the V beta regions. (See e.g., Muller, 1998, Structure 6:1153-
1167).
In one embodiment, a polynucleotide encoding a modified TCR, or an
antigen-binding fragment thereof, is provided. In other related embodiments, the
polynucleotide may be a variant of a cleotide encoding the modified TCR.
Polynucleotide variants may have substantial identity to a polynucleotide sequence
encoding a modified TCR described herein. For example, a polynucleotide may be a
polynucleotide comprising at least 70% sequence ty, preferably at least 75%,
80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or , sequence identity
compared to a reference polynucleotide sequence such as a sequence ng an
TCR described herein, using the methods described herein, (e.g., BLAST analysis
using standard parameters, as described below). One skilled in this art will recognize
that these values can be appropriately adjusted to determine corresponding identity
of proteins encoded by two nucleotide sequences by taking into account codon
degeneracy, amino acid similarity, reading frame positioning and the like.
Typically, cleotide variants will contain one or more substitutions,
additions, deletions and/or insertions, ably such that the binding affinity of the
TCR encoded by the variant polynucleotide is not ntially diminished relative to
an TCR encoded by a polynucleotide sequence specifically set forth herein.
When comparing polynucleotide sequences, two sequences are said to be
ical” if the sequence of nucleotides in the two sequences is the same when
aligned for m correspondence, as bed below. Comparisons between
two sequences are typically performed by comparing the sequences over a
comparison window to fy and compare local regions of sequence similarity. A
“comparison window” as used herein, refers to a segment of at least about 20
contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence
may be compared to a reference sequence of the same number of contiguous
positions after the two sequences are optimally aligned.
] Optimal ent of sequences for comparison may be conducted using
the Megalign program in the Lasergene suite of ormatics software (DNASTAR,
Inc., Madison, WI), using default parameters. This program embodies several
alignment schemes described in the following references: Dayhoff, MO. (1978) A
model of evolutionary change in proteins — Matrices for detecting distant
relationships. In Dayhoff, M.O. (ed.) Atlas of Protein Sequence and Structure,
National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-
358; Hein J., Unified Approach to Alignment and enes, pp. 626-645 (1990);
Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins,
D.G. and Sharp, P.M., CABIOS 5:151—153 (1989); Myers, E.W. and Muller W.,
CABIOS 7 (1988); Robinson, E.D., Comb. Theor 11:105 (1971); Santou, N.
Nes, M., Mol. Biol. Evol. 4:406-425 (1987); Sneath, P.H.A. and Sokal, R.R.,
Numerical Taxonomy — the Principles and Practice of Numerical Taxonomy,
Freeman Press, San Francisco, CA (1973); Wilbur, W.J. and Lipman, D.J., Proc.
Natl. Acad., Sci. USA 80:726-730 (1983).
Alternatively, optimal alignment of sequences for comparison may be
conducted by the local identity algorithm of Smith and Waterman, Add. APL. Math
2:482 (1981), by the identity alignment thm of Needleman and Wunsch, J. Mol.
Biol. 482443 (1970), by the search for similarity methods of Pearson and Lipman,
Proc. Natl. Acad. Sci. USA 85: 2444 (1988), by computerized implementations of
these algorithms (GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin
Genetics re Package, Genetics Computer Group (GCG), 575 Science Dr.,
Madison, WI), or by tion.
One preferred example of algorithms that are suitable for determining
percent ce identity and sequence similarity are the BLAST and BLAST 2.0
algorithms, which are described in Altschul et al., Nucl. Acids Res. 25:3389-3402
(1977), and Altschul et al., J. Mol. Biol. 3-410 (1990), respectively. BLAST
and BLAST 2.0 can be used, for example with the parameters described herein, to
determine t sequence identity among two or more the cleotides.
Software for performing BLAST analyses is publicly ble through the National
Center for hnology Information. In one illustrative example, cumulative scores
can be calculated using, for nucleotide sequences, the parameters M (reward score
for a pair of matching residues; always >0) and N (penalty score for mismatching
residues; always <0). ion of the word hits in each direction are halted when:
the tive alignment score falls off by the quantity X from its maximum achieved
value; the cumulative score goes to zero or below, due to the accumulation of one or
more negative-scoring residue alignments; or the end of either sequence is reached.
The BLAST algorithm parameters W, T and X ine the sensitivity and speed of
the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a
ngth (W) of 11, and expectation (E) of 10, and the BLOSUM62 scoring matrix
(see Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1989)) alignments,
(B) of 50, expectation (E) of 10, M=5, N=-4 and a comparison of both strands.
In certain embodiments, the “percentage of sequence identity” is
determined by comparing two optimally d sequences over a window of
comparison of at least 20 positions, wherein the portion of the cleotide
sequence in the comparison window may comprise additions or deletions (i.e., gaps)
of 20 percent or less, y 5 to 15 percent, or 10 to 12 percent, as compared to
the reference sequences (which does not comprise additions or deletions) for
optimal alignment of the two sequences. The percentage is calculated by
determining the number of positions at which the identical nucleic acid bases occurs
in both ces to yield the number of matched positions, dividing the number of
matched positions by the total number of positions in the nce sequence (i.e.,
the window size) and multiplying the results by 100 to yield the percentage of
sequence identity.
It will be appreciated by those of ordinary skill in the art that, as a result of
the degeneracy of the genetic code, there are many nucleotide sequences that
encode an TCR as described herein. Some of these polynucleotides bear minimal
sequence identity to the nucleotide sequence of the native or original polynucleotide
sequence that encode modified TCRs that bind to, e.g., the same antigen.
Nonetheless, polynucleotides that vary due to differences in codon usage are
sly contemplated by the present disclosure. In certain embodiments,
ces that have been codon-optimized for ian expression are
specifically contemplated.
Standard techniques for cloning, DNA isolation, amplification and
purification, for enzymatic reactions involving DNA ligase, DNA polymerase,
restriction endonucleases and the like, and various tion techniques are those
known and ly employed by those skilled in the art. A number of standard
techniques are described in Sambrook et al. (1989) Molecular Cloning, Second
Edition, Cold Spring Harbor Laboratory, Plainview, New York; Maniatis et al. (1982)
Molecular Cloning, Cold Spring Harbor Laboratory, Plainview, New York; Wu (ed.)
(1993) Meth. Enzymol. 218, Part I; Wu (ed.) (1979) Meth Enzymol. 68; Wu et al.
(eds.) (1983) Meth. Enzymol. 100 and 101; an and Moldave (eds.) Meth.
Enzymol. 65; Miller (ed.) (1972) Experiments in Molecular cs, Cold Spring
Harbor Laboratory, Cold Spring Harbor, New York; Old and Primrose (1981)
Principles of Gene Manipulation, University of California Press, Berkeley; Schleif and
Wensink (1982) Practical Methods in Molecular Biology; Glover (ed.) (1985) DNA
Cloning Vol. I and II, IRL Press, Oxford, UK; Hames and Higgins (eds.) (1985)
Nucleic Acid Hybridization, IRL Press, , UK; and Setlow and Hollaender
(1979) Genetic Engineering: ples and Methods, Vols. 1-4, Plenum Press, New
York. Abbreviations and nomenclature, where employed, are deemed standard in
the field and commonly used in professional journals such as those cited herein.
Homology between nucleotide sequences can be determined by DNA
hybridization analysis, wherein the stability of the double-stranded DNA hybrid is
dependent on the extent of base pairing that occurs. Conditions of high temperature
and/or low salt content reduce the stability of the hybrid, and can be varied to
prevent annealing of sequences having less than a ed degree of homology.
For instance, for sequences with about 55% G - C content, hybridization and wash
conditions of 40 - 50°C, 6 X SSC (sodium chloride/sodium citrate buffer) and 0.1%
SDS (sodium dodecyl e) indicate about 60 — 70% homology, hybridization and
wash conditions of 50 - 65°C, 1 X SSC and 0.1% SDS indicate about 82 - 97%
homology, and hybridization and wash conditions of 52°C, 0.1 X SSC and 0.1% SDS
indicate about 99 — 100% homology. A wide range of computer programs for
comparing nucleotide and amino acid sequences (and measuring the degree of
homology) are also available, and a list providing sources of both commercially
ble and free software is found in l et al. . Readily available
sequence comparison and multiple sequence alignment algorithms are, respectively,
the Basic Local Alignment Search Tool ) (Altschul et al., 1997) and ClustalW
programs. BLAST is available on the Internet at ncbi.n|m.nih.gov and a version of
ClustalW is available at www2.ebi.ac.uk.
Industrial strains of microorganisms (e.g., Aspergillus niger, Aspergillus
ficuum, Aspergillus awamori, Aspergillus , Trichoderma reesei, Mucor miehei,
romyces lactis, Pichia pastoris, Saccharomyces cerevisiae, Escherichia coli,
Bacillus subtilis or Bacillus licheniformis), insect (Drosophila), mammalian
(e.g.,Chinese hamster ovary cell lines, CHO), or plant species (e.g., ,
soybean, corn, potato, barley, rye, wheat) may be used as host cells for the
recombinant production of the TCR proteins. In certain embodiments, the first step in
the heterologous expression of a high affinity TCR protein or soluble protein, an
expression construct is assembled to include the TCR or soluble TCR coding
ce and l sequences such as promoters, enhancers and ators.
Other sequences such as signal sequences and selectable s may also be
included. To achieve extracellular expression of the TCR, the expression construct
may include a secretory signal ce. In embodiments, the signal sequence is
not included on the expression construct if cytoplasmic expression is desired. In
embodiments, the promoter and signal sequence are functional in the host cell and
provide for expression and secretion of the TCR or soluble TCR protein.
Transcriptional terminators may be included to ensure efficient transcription.
Ancillary sequences enhancing expression or protein purification may also be
included in the expression construct.
Various promoters (transcriptional initiation regulatory region) may be used
according to the disclosure. The selection of the appropriate promoter may be
dependent upon the proposed expression host. Promoters from heterologous
sources may be used as long as they are onal in the chosen host.
Promoter selection is also dependent upon the desired efficiency and level
of peptide or protein production. Inducible promoters such as tac are often ed
in order to dramatically increase the level of protein sion in E. coli.
Overexpression of proteins may be harmful to the host cells. Consequently, host cell
growth may be limited. The use of inducible er s allows the host cells
to be ated to acceptable densities prior to induction of gene expression, thereby
facilitating higher product yields.
] Various signal sequences may be used according to the disclosure. A
signal sequence which is homologous to the TCR coding sequence may be used.
Alternatively, a signal sequence which has been selected or designed for efficient
secretion and processing in the expression host may also be used. For example,
suitable signal sequence/host cell pairs include the B. subtilis sacB signal sequence
for secretion in B. subtilis, and the Saccharomyces cerevisiae o-mating factor or P.
pastoris acid phosphatase phol signal sequences for P. pastoris secretion. The
signal sequence may be joined directly through the sequence encoding the signal
peptidase cleavage site to the protein coding sequence, or through a short
nucleotide bridge consisting of usually fewer than ten codons, where the bridge
ensures correct reading frame of the downstream TCR sequence.
ts for enhancing ription and translation have been identified
for eukaryotic protein expression systems. For example, positioning the cauliflower
mosaic virus (CaMV) promoter 1000 bp on either side of a heterologous er
may elevate transcriptional levels by 10- to 400-fold in plant cells. The expression
construct should also include the riate translational initiation sequences.
Modification of the sion construct to include a Kozak consensus sequence for
proper translational initiation may increase the level of translation by 10 fold.
A selective marker is often employed, which may be part of the expression
construct or separate from it (e.g., carried by the expression vector), so that the
marker may integrate at a site different from the gene of interest. Examples include
markers that confer ance to antibiotics (e.g., bla confers resistance to ampicillin
for E. coli host cells, nptll confers kanamycin resistance to a wide variety of
prokaryotic and eukaryotic cells) or that permit the host to grow on minimal medium
(e.g., HIS4 enables P. pastoris or His- S. cerevisiae to grow in the absence of
histidine). The selectable marker has its own transcriptional and translational
initiation and termination regulatory regions to allow for ndent expression of
the marker. If otic resistance is employed as a , the tration of the
antibiotic for selection will vary depending upon the antibiotic, generally ranging from
to 600 ug of the otic/mL of medium.
The sion construct is assembled by ing known recombinant
DNA techniques (Sambrook et al., 1989; Ausubel et al., 1999). Restriction enzyme
digestion and ligation are the basic steps employed to join two fragments of DNA.
The ends of the DNA fragment may e modification prior to ligation, and this
may be accomplished by filling in overhangs, deleting terminal portions of the
fragment(s) with nucleases (e.g., Exolll), site directed nesis, or by adding
new base pairs by PCR. Polylinkers and adaptors may be employed to facilitate
joining of selected fragments. The expression construct is typically assembled in
stages employing rounds of restriction, ligation, and transformation of E. coli.
Numerous cloning vectors suitable for construction of the sion construct are
known in the art (AZAP and pBLUESCRIPT SK-1, Stratagene, LaJoIla, CA; pET,
Novagen Inc., Madison, WI - cited in Ausubel et al., 1999) and the particular choice
is not critical to the disclosure. The selection of cloning vector will be influenced by
the gene transfer system selected for introduction of the expression construct into
the host cell. At the end of each stage, the ing construct may be analyzed by
restriction, DNA sequence, hybridization and PCR analyses.
The expression construct may be transformed into the host as the cloning
vector construct, either linear or ar, or may be removed from the cloning vector
and used as is or introduced onto a delivery vector. The delivery vector facilitates the
introduction and maintenance of the expression construct in the selected host cell
type. The expression construct is introduced into the host cells by any of a number of
known gene transfer systems (e.g., l ence, chemically mediated
ormation, protoplast transformation, electroporation, biolistic transformation,
transfection, or conjugation) (Ausubel et al., 1999; Sambrook et al., 1989). The gene
transfer system selected depends upon the host cells and vector systems used.
For instance, the expression construct can be introduced into S. siae
cells by protoplast transformation or electroporation. Electroporation of S. cerevisiae
is readily accomplished, and yields ormation efficiencies comparable to
spheroplast transformation.
Monoclonal or onal antibodies, preferably monoclonal, specifically
reacting with a TCR protein at a site other than the ligand binding site may be made
by methods known in the art, and many are commercially available. See, e.g.,
Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor
Laboratories; Goding (1986) onal Antibodies: Principles and Practice, 2d ed.,
Academic Press, New York; and Ausubel et al. (1999) Current ols in Molecular
Biology, John Wiley & Sons, Inc, New York.
] TCRs in cell-bound or e form which are specific for a particular target
ligand are useful, for example, as diagnostic probes for screening biological samples
(such as cells, tissue samples, biopsy material, bodily fluids and the like) or for
detecting the presence of the target ligand in a test . Frequently, the TCRs
are labeled by joining, either covalently or noncovalently, a nce which
provides a detectable signal. le labels include but are not limited to
uclides, enzymes, substrates, cofactors, inhibitors, fluorescent agents,
chemiluminescent , magnetic particles and the like. Additionally the TCR can
be coupled to a ligand for a second binding molecule: for example, the TCR can be
biotinylated. Detection of the TCR bound to a target cell or molecule can then be
effected by binding of a detectable streptavidin (a streptavidin to which a fluorescent,
radioactive, chemiluminescent, or other detectable molecule is ed or to which
an enzyme for which there is a chromophoric substrate ble). United States
Patents describing the use of such labels and/or toxic compounds to be covalently
bound to the scTCR include but are not limited to Nos. 3,817,837; 3,850,752;
3,927,193; 3,939,350; 3,996,345; 437; 4,275,149; 4,331,647; 4,348,376;
4,361,544; 4,468,457; 4,444,744; 4,640,561; 4,366,241; RE 35,500; 5,299,253;
,101,827; 5,059,413.
Labeled TCRs can be detected using a monitoring device or method
appropriate to the label used. Fluorescence microscopy or fluorescence activated
cell sorting can be used where the label is a fluorescent moiety, and where the label
is a radionuclide, gamma counting, autoradiography or liquid scintillation counting,
for example, can be used with the proviso that the method is appropriate to the
sample being analyzed and the radionuclide used. In addition, there can be
secondary detection molecules or particle employed where there is a detectable
molecule or particle which recognized the portion of the TCR which is not part of the
binding site for the target ligand in the absence of a MHC component as noted
herein. The art knows useful compounds for diagnostic imaging in situ; see, e.g.,
U.S. Patent No. 827; 5,059,413. Radionuclides useful for therapy and/or
imaging in vivo include 111indium, 97Rubidium, 125iooiine, 131Iodine, 123Iodine,
67Gallium, 99Technetium. Toxins include diphtheria toxin, ricin and castor bean toxin,
among others, with the proviso that once the TCR-toxin complex is bound to the cell,
the toxic moiety is alized so that it can exert its cytotoxic effect. lmmunotoxin
technology is well known to the art, and suitable toxic molecules include, without
limitation, chemotherapeutic drugs such as vindesine, antifolates, e.g., methotrexate,
cisplatin, cin, anthrocyclines such as ycin, daunorubicin or
adriamycin, and cytotoxic proteins such as me inactivating ns (e.g.,
diphtheria toxin, pokeweed antiviral protein, abrin, ricin, pseudomonas exotoxin A or
their recombinant derivatives. See, lly, e.g., Olsnes and Pihl (1982) Pharmac.
Ther. 25:355-381 and Monoclonal Antibodies for Cancer Detection and y,
Eds. Baldwin and Byers, pp. 159-179, Academic Press, 1985.
] The general structure of TCR molecules and methods of making and
using, including binding to a peptide:Major Histocompatibility Complex have been
disclosed. See, for example PCT/U898/04274; PCT/U898/20263; W099/60120.
ceutical Compositions and Therapeutic Agents
TCRs specific for a particular target ligand are useful in treating animals
and mammals, including humans believed to be suffering from a disease associated
with the particular antigen, e.g., a neoplastic disease or disorder, such as cancer.
Examples of types of cancers that may be treated according to the methods
described herein include, but are not limited to, Wilm’s tumour, r cancer,
breast cancer, colon cancer, colorectal cancer, esophageal carcinomas, gastric
cancer, hepatocellular carcinoma, kidney cancer, leukemia, liver cancer, lung
cancer, lymphoma, melanoma, neuroblastoma, non-small cell lung carcinoma, oral
cancer, osteosarcoma, ovarian cancer, atic cancer, prostate cancer, renal
cancer, skin cancer, small cell lung carcinoma, and testicular cancer.
Therapeutic products can be made using the materials shown herein.
Effective amounts of therapeutic ts are the minimum dose that produces a
measurable effect in a subject. Therapeutic products are easily prepared by one of
ordinary skill in the art. In one embodiment, a scTCR of the disclosure is
administered directly to a patient. In one embodiment, a scTCR of the disclosure is
linked to PEG or to immunoglobulin constant regions, as known in the art. This
embodiment lengthens the serum clearance. In one embodiment, the scTCR is
linked to a chemotherapeutic agent or drug in order to r the drug to a target cell
such as a cancer cell. In one embodiment, the scTCR is linked to a biologic effector
le such as a cytokine (Tayal and Kalra (2008) EurJ Pharmacol, 579, 1-12). In
one embodiment, the scTCR is linked to a cytokine with umor activity, such as
lL-2, lL-12, or TNFd (Wong et al. (2011) n Eng Des Sel, 24, 373—83). In one
embodiment, the scTCR is linked to an -inhibitory cytokine, such as lL—10 or
lL-13 (Stone et al. (2012) Protein Engineering). In one ment, the scTCR is
linked to another n g molecule to form a bispecific agent (Miller et al.
(2010) Protein Eng Des Sel, 23, 549-57; Thakur and Lum (2010) Curr Opin Mol
Ther, 12, 340-9). In one embodiment, the bispecific molecule is comprised of a
scTCR linked to a single chain Fv, such as an anti-CD3 ((Bargou et al. (2008)
Science, 321, 974—7; Liddy et al. (2012) Nat Med, 18, 980-7), to crosslink T cells and
diseased cells. In one embodiment, the scTCR is linked to TCR signaling domains,
such as CD3, to form a chimeric antigen receptor ((Porter et al. (2011) N Engl J Med,
365, 725-33; Sadelain et al. (2009) Curr Opin Immunol, 21, 215-23; Stroncek et al.
(2012) J Transl Med, 10, 48). These methods and other methods of administering,
such as enously, are known in the art. Useful dosages can be determined by
one of ry skill in the art.
The scTCR itions can be formulated by any of the means known in
the art. They can be typically prepared as injectables, especially for intravenous,
intraperitoneal or synovial administration (with the route determined by the particular
e) or as formulations for asal or oral administration, either as liquid
solutions or suspensions. Solid forms suitable for solution in, or suspension in, liquid
prior to injection or other administration may also be prepared. The preparation may
also, for example, be emulsified, or the protein(s)/peptide(s) encapsulated in
liposomes.
] The active ingredients are often mixed with optional pharmaceutical
additives such as excipients or carriers which are pharmaceutically acceptable and
compatible with the active ingredient. Suitable ents include but are not d
to water, saline, dextrose, glycerol, ethanol, or the like and ations f.
The concentration of the scTCR in injectable, aerosol or nasal formulations is usually
in the range of 0.05 to 5 mg/ml. The selection of the particular effective dosages is
known and performed without undue experimentation by one of ordinary skill in the
art. Similar dosages can be administered to other mucosal surfaces.
In addition, if desired, vaccines that could e a scTCR may contain
minor amounts of pharmaceutical ves such as auxiliary substances such as
wetting or emulsifying agents, pH buffering agents, and/or adjuvants which enhance
the effectiveness of the vaccine. Examples of adjuvants which may be effective
include but are not limited to: aluminum hydroxide; N—acetyl-muramyl-L-threonyl-
Disoglutamine (thr-MDP); N-acetyl-nor—muramyl-L-alanyl-D-isoglutamine (CGP
11637, referred to as nor-MDP); N—acetylmuramyl—L-alanyl—D-isoglutaminyl-L-
alanine(1'-2'-dipalmitoyl-sn-glycero-3hydroxyphosphoryloxy)-ethylamine (CGP
19835A, referred to as MTP-PE); and RIBI, which ns three components
extracted from bacteria: osphoryl lipid A, trehalose dimycolate and cell wall
skeleton (MPL+TDM+CWS) in a 2% squalene/Tween® 80 emulsion. Such additional
formulations and modes of administration as are known in the art may also be used.
The scTCRs of the present disclosure and/or binding fragments having
primary structure similar (more than 90% identity) to the TCR variable regions and
which maintain the high affinity for the target ligand may be formulated into vaccines
as neutral or salt forms. Pharmaceutically acceptable salts include but are not limited
to the acid addition salts (formed with free amino groups of the peptide) which are
formed with inorganic acids, e.g., hydrochloric acid or phosphoric acids; and c
acids, e.g., acetic, oxalic, tartaric, or maleic acid. Salts formed with the free carboxyl
groups may also be derived from inorganic bases, e.g., sodium, potassium,
ammonium, calcium, or ferric hydroxides, and c bases, e.g., isopropylamine,
trimethylamine, 2—ethylamino-ethanol, histidine, and procaine.
] scTCRs for therapeutic use are administered in a manner compatible with
the dosage formulation, and in such amount and manner as are lactically
and/or therapeutically effective, according to what is known to the art. The quantity to
be stered, which is generally in the range of about 100 to 20,000 ug of n
per dose, more generally in the range of about 1000 to 10,000 ug of protein per
dose. Similar compositions can be administered in similar ways using labeled
scTCRs for use in imaging, for e, to detect cells to which a target ligand is
bound. Precise amounts of the active ingredient required to be administered may
depend on the judgment of the physician or veterinarian and may be peculiar to each
individual, but such a ination is within the skill of such a practitioner.
The TCR product may be given in a single dose; two dose schedule, for
example two to eight weeks apart; or a multiple dose schedule. A multiple dose
schedule is one in which a primary course of treatment may include 1 to 10 or more
separate doses, ed by other doses administered at subsequent time intervals
as ed to maintain and/or reinforce the response.
Every formulation or combination of ents described or exemplified
can be used to practice the disclosure, unless othenNise stated. Specific names of
nces are intended to be exemplary, as it is known that one of ordinary skill in
the art can name the same substances differently. When a compound is described
herein such that a particular isomer or omer of the compound is not specified,
for example, in a formula or in a chemical name, that description is intended to
include each isomers and enantiomer of the compound described individual or in any
ation. One of ordinary skill in the art will appreciate that methods, target
ligands, biologically active groups, starting materials, and synthetic methods other
than those specifically exemplified can be ed in the practice of the disclosure
without resort to undue experimentation. All art-known functional equivalents, of any
such methods, target ligands, biologically active groups, ng materials, and
synthetic methods are intended to be included in this disclosure. Whenever a range
is given in the specification, for example, a temperature range, a time range, or a
composition range, all intermediate ranges and subranges, as well as all individual
values included in the ranges given are intended to be included in the disclosure.
The exact formulation, route of administration and dosage can be chosen
by the individual physician in view of the patient's condition (see e.g., Fingl et. al., in
The Pharmacological Basis of Therapeutics, 1975, Ch. 1 p. 1).
It should be noted that the attending physician would know how to and
when to ate, interrupt, or adjust administration due to toxicity, or to organ
dysfunctions. Conversely, the attending ian would also know to adjust
treatment to higher levels if the clinical response were not adequate (precluding
toxicity). The magnitude of an administered dose in the management of the disorder
of interest will vary with the severity of the condition to be treated and to the route of
administration. The severity of the condition may, for example, be evaluated, in part,
by standard prognostic evaluation methods. Further, the dose and perhaps dose
ncy, will also vary according to the age, body weight, and se of the
individual patient. A program comparable to that discussed above also may be used
in veterinary ne.
Depending on the specific conditions being treated and the targeting
method selected, such agents may be formulated and administered systemically or
y. Techniques for formulation and administration may be found in Alfonso and
Gennaro (1995). Suitable routes may include, for example, oral, rectal, transdermal,
vaginal, transmucosal, or intestinal administration; parenteral delivery, including
uscular, aneous, or intramedullary injections, as well as intrathecal,
intravenous, or eritoneal injections.
For injection, the agents of the disclosure may be formulated in s
solutions, preferably in logically compatible buffers such as Hanks' solution,
Ringer's solution, or logical saline buffer. For transmucosal administration,
penetrants appropriate to the barrier to be permeated are used in the formulation.
Such penetrants are generally known in the art.
Use of pharmaceutically acceptable carriers to formulate the compounds
herein disclosed for the practice of the disclosure into dosages suitable for systemic
stration is within the scope of the disclosure. With proper choice of carrier and
suitable cturing practice, the compositions of the present disclosure, in
particular those formulated as solutions, may be administered parenterally, such as
by intravenous injection. Appropriate compounds can be ated readily using
pharmaceutically acceptable carriers well known in the art into dosages suitable for
oral administration. Such carriers enable the compounds of the disclosure to be
formulated as s, pills, capsules, liquids, gels, , slurries, suspensions and
the like, for oral ingestion by a patient to be treated.
] Agents intended to be administered intracellularly may be administered
using techniques well known to those of ordinary skill in the art. For example, such
agents may be encapsulated into liposomes, and then administered as described
above. Liposomes are spherical lipid rs with aqueous interiors. All molecules
present in an aqueous on at the time of liposome formation are incorporated
into the aqueous interior. The liposomal contents are both protected from the
external microenvironment and, because liposomes fuse with cell membranes, are
efficiently delivered into the cell cytoplasm. onally, due to their hydrophobicity,
small organic molecules may be directly administered intracellularly.
Pharmaceutical compositions suitable for use in the t disclosure
include compositions wherein the active ingredients are contained in an effective
amount to achieve the intended purpose. ination of the effective amounts is
well within the capability of those skilled in the art, especially in light of the detailed
disclosure provided herein.
In addition to the active ingredients, these pharmaceutical compositions
may n suitable pharmaceutically acceptable carriers comprising excipients and
aries which facilitate processing of the active nds into preparations
which can be used pharmaceutically. The preparations formulated for oral
administration may be in the form of tablets, dragees, capsules, or solutions,
including those formulated for delayed release or only to be ed when the
pharmaceutical reaches the small or large intestine.
The pharmaceutical itions of the t disclosure may be
manufactured in a manner that is itself known, e.g., by means of conventional
mixing, dissolving, granulating, dragee-making, levitating, emulsifying,
encapsulating, ping or lyophilizing processes.
Pharmaceutical formulations for parenteral administration include aqueous
solutions of the active compounds in water-soluble form. Additionally, suspensions of
the active compounds may be prepared as appropriate oily injection suspensions.
Suitable lipophilic ts or vehicles include fatty oils such as sesame oil, or
tic fatty acid esters, such as ethyl oleate or triglycerides, or mes.
Aqueous injection suspensions may contain substances which increase the viscosity
of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
Optionally, the suspension may also contain le stabilizers or agents which
se the solubility of the compounds to allow for the preparation of highly
concentrated solutions.
Pharmaceutical preparations for oral use can be obtained by combining
the active compounds with solid excipient, optionally grinding a resulting mixture,
and processing the mixture of es, after adding suitable auxiliaries, if desired, to
obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as
sugars, including lactose, sucrose, ol, or sorbitol; cellulose preparations such
as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum
tragacanth, methyl cellulose, ypropylmethyl-cellulose, sodium
carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If d, disintegrating
agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic
acid or a salt thereof such as sodium te.
Dragee cores are provided with le coatings. For this purpose,
concentrated sugar solutions may be used, which may optionally n gum arabic,
talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide,
lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or
pigments may be added to the tablets or dragee coatings for identification or to
characterize different combinations of active compound doses.
Pharmaceutical preparations which can be used orally include push-fit
capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a
plasticizer, such as glycerol or sorbitol. The push—fit capsules can n the active
ingredients in admixture with filler such as lactose, binders such as starches, and/or
lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft
capsules, the active compounds may be dissolved or suspended in suitable liquids,
such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers
may be added.
Methods of Treatment
The high affinity TCRs and pharmaceutical itions comprising a
high affinity TCR may be used, for e, to treat a patient having a cancer,
tumor, malignancy, or stic disease or er. In one embodiment, a method
of treating a patient having cancer comprises administering a high affinity TCR
described herein. In one embodiment, the high affinity TCR is specific for WT1. In
one embodiment, the TCR comprises a VB comprising the amino acid sequence set
forth in SEQ ID NO:3. In r ment, the TCR comprises a V0I sing
the amino acid sequence set forth in SEQ ID NO:4. In one embodiment, the high
affinity TCR is a single chain TCR comprising the amino acid sequence set forth in
SEQ ID NO:5. In another embodiment, the high affinity TCR is administered in
combination with a therapeutic agent, e.g., a chemotherapeutic agent. In yet another
embodiment, the high affinity TCR is conjugated to a biologically active group.
] Another aspect of the invention provides a method for the adoptive
transfer of T cells to a patient in need thereof, comprising administering T cells that
s either the wild type TCR or a high affinity TCR described herein. In one
embodiment, the T cells have been transfected with a polynucleotide that encodes a
TCR that is specific for WT1. In one embodiment, the TCR comprises a VB
comprising the amino acid sequence set forth in SEQ ID NO:3. In another
embodiment, the TCR comprises a Va comprising the amino acid sequence set forth
in SEQ ID NO:4. In one embodiment, the high affinity TCR is a single chain TCR
comprising the amino acid sequence set forth in SEQ ID NO:5.
EXAMPLES
The following es further describe non-limiting examples of the
disclosure.
EXAMPLE 1
ENGINEERING TCRs FOR HIGHER AFFINITY FOR E/HLA-A2 ANTIGENS
The general strategy used to discover, or te single-chain TCRs for
ed affinity and stability is shown in Figure 3. The process involves six steps,
as illustrated:
1) Cloning the Va and V8 TCR genes from a T cell clone such as P22
(sequence shown in Figure 1, and the VB comprising the amino acid sequence set
forth in SEQ ID NO:1 and the Va comprising the amino acid sequence set forth in
SEQ ID NO:2) as a single chain TCR format for display. The V regions recognize the
HLA-A2-restricted antigenic peptide WT1 (SEQ ID NO:6) in complex with . In
the present invention, the TCR V region genes from clone P22 (e.g. Dossett et al.
(2009) Mol Ther. 17(4), 742) were cloned as a single-chain format (VB-linker-Va) and
introduced into a yeast display vector for sion on the surface of yeast (Figure
2) tion of an error prone library and FACs or magnetic bead
selection for stabilized variants with an anti-V8 antibody. e the single-chain
Va and V8 TCRs are often unstable due to loss of the stabilizing constant regions,
error—prone mutagenesis libraries are generated to select for izing mutations
that allow for stable expression on the surface of yeast, although other display
formats including but not limited to phage and mammalian display may be used.
Phage display vectors and cloning have yielded y sizes of 1011, whereas yeast
display vectors and homologous recombination steps have yielded library sizes of
101° ((Benatuil et al. (2010) Protein Eng Des Sel, 23, 155-9). Various methods have
been used for selection of variants, including affinity-based binding to immobilized
ligands (phage display) or magnetic particle ions with antigens (yeast display),
or scent activated cell sorting with d-peptide-MHC antigens (yeast
display). Utilizing antibodies against the TCR V8 that recognize folded epitopes,
fluorescent activated cell sorting (FACS) or magnetic bead selection are used to
isolate variants with improved dy binding in the present example.
3) scTCR clones isolated from the selection of the error prone library are
assessed for thermal stability and a ize variant is chosen for a template for
affinity maturation, and ced. Typically, single-site mutations are identified that
contribute to increased surface levels on yeast, and greater stability in solution.
4) The ized scTCR sequences are used as a template for the
generation of CDR libraries, usually in the CDR1a, CDR3a, CDR38, although other
regions including but not limited to the CDR1B, , CDR2B, and HV4 can also
be used. In the t disclosure, yeast displayed variants are selected for
improved binding to peptide:MHC, from the CDR libraries, by using magnetic bead
selections and/or scence activated cell sorting (FACS), although selections
ing other methods ing but not limited to panning with phage display or
magnetic ions or FACS with ian display may be used.
5) scTCR clones isolated from the selection of the CDR ies are
assessed for ic binding to the peptide:MHC against which they were
engineered. Plasmids are rescued from the yeast clones, and sequenced.
6) If further improvements of affinity required, the scTCR clone selected in
step 5 can be used as a template for the generation of additional libraries in other
loops or regions that did not select mutations such as CDR1(X, CDR30i, CDR3B,
although other s including but not limited to the CDR1B, CDRZOL, CDR2B, and
HV4 can also be used. Examples of each of these steps are described further below.
EXAMPLE 2
ANALYSIS OF THE HUMAN TCR A6, WHICH USES THE Va2, IN X WITH TAX:HLA.A2
TCRs all adopt a similar lg-fold and docking angle, and TCR recognition of
pepMHC is mediated entirely by specific residues on CDR loops a et al. (2009)
Nat Immunol, 10, 143-7; Marrack et al. (2008) Annu Rev Immunol, 26, 171-203;
Rudolph et al. (2006) Annu Rev Immunol, 24, 419-66)). Although crystal structures
for the WT1 TCRs are not available at the time of the present disclosure, the
structure of the A6:Tax peptide:HLA-A2 complex (PDB: 1AO7) (Garboczi et al.
(1996) Nature, 384, 134-141), which used the same Va2 domain of the WT1 P22
TCR is shown. The side view of the complex showed that the ends of the variable
domains that contained the six CDRs docked onto the Tax:HLA.A2 molecule, with
the central region of the binding site positioned over the peptide Tax (Figure 2A).
The crystal structure does not include the constant region or, although the constant
regions help stabilize the full length construct. Stabilizing mutations selected in step
2 described above are often selected in framework regions, such as the VOJVB
interface or where the junctions of the Cor/Von or CB/VB interface occurs in the full
length TCR.
The top down view of the TaX:HLA-A2 complex, with the TCR “removed”,
except for the six CDR loops is shown (Figure 28). This view shows that the TCR
adopts a diagonal position over the peptide-MHC, a finding which has now been
observed for all TCRzpeptide-MHC structures. In this orientation, the two CDR3
loops are positioned over the peptide, while there are s residues from CDR1
and CDR2 loops that interact predominantly with the helices of the MHC molecule.
For purposes of ty maturation in steps 4 and 6, these loops are often the
ed for the generation of affinity maturations ies, although other regions
may be used.
EXAMPLE 3
YEAST DISPLAY OF WT1 TCRS
In order to perform selections for improved stability (step 2) or improved
affinity (step 5), it is ary to use a display system in which a library of TCR
mutants can be screened for binding to an antibody which recognizes a conformation
epitope or a peptide:MHC ligand, respectively. Three display systems have been
used for engineering TCRs for higher affinity, and could be used for this process:
yeast display, phage display, and T cell (mammalian cell) display. Alternative y
methods, such as ribosome, RNA, DNA, and CIS display, may also be suitable for
this process. In all of these cases, the wild type TCR with low ty for the antigen
was cloned into the , and used as a template for engineering TCRs with
enhanced stability and affinity against the peptidezMHC ligand. Any of these systems
could be applied to the approach described here, in which a single TCR is used as a
template for libraries and the selection of TCRs with enhanced binding properties.
] In the present example, yeast display was used as the platform (Figure 4).
The WT1 TCR was used as the template for stabilizing mutations via error prone
mutagenesis, and stabilized clones ed from the selections were used as
templates for affinity maturation.
EXAMPLE 4
ERROR-PRONE Y CONSTRUCTION AND SELECTION OF A STABILIZED WT1 TCR, WT1-
] The WT1 error-prone y was generated as previously described
(Richman et al. (2009) Methods Mol Biol 504, 323-350) utilizing the WT1-reactive
cell line called P22 as a template. The human WT1 prone scTCR library was
thus introduced into the yeast display vector by combining the linearized pCT302
vector, WT1 error-prone PCR product, and competent EBY100 yeast cells. The
resultant library contained about 2.3 X 107 independent clones, was judged by
plating limiting dilution aliquots of yeast after electroporation. The library was
selected for binding to two antibodies that recognize human hV[33, anti-hV[33.1 FITC
lgG (Thermo Scientific) and anti-hVBB FITC lgM (Beckman Coulter), via FACS
according to Table 1.
Table 1. Sorting ions
Thermo Scientific hVB3.1 FITC (1 :10); AlexaFluor® 647 Goat anti—mouse lgG
(1:100)
Beckman Coulter hVB3 FITC lgM (1 :10); Goat anti-mouse lgM APC (1 :4)
Thermo Scientific hVB3.1 FITC (1:10); luor 647® Goat anti-mouse lgG
(1:100)
Beckman Coulter hVB3 FITC lgM (1:10); Goat anti-mouse lgM APC (1:4)
Thermo Scientific hVB3.1 FITC (1:10); AlexaFluor® 647 Goat anti-mouse lgG
(1:100)
n Coulter hVB3 FITC lgM (1:10); Goat ouse lgM APC (1:4)
Using thermal denaturation s, we have identified these antibodies to
recognize folded epitopes on V03 (data not shown). Signals were amplified using
AlexaFluor® 647 Goat anti-mouse lgG (Life Technologies) and Goat anti-mouse lgM
APC (lnvitrogen) secondary antibodies. During 3 iterative sorts, a VB3-positively
staining population emerged (Figure 5A). ing the 3rd sort, a clone called WT1-
D13 was isolated for improved V[33 fluorescence (Figure SB) and showed thermal
stability when heated to 80 °C (data not shown). The WT1-D13 clone was used as a
template for affinity maturation.
EXAMPLE 5
CDR10I LIBRARY CONSTRUCTION AND SELECTION OF A WT1 TCR WITH ENHANCED BINDING
TO WT1:HLA.A2, WT1.1
] The stabilized 3 clone isolated from selection of error-prone PCR
ies was used as a template for generation of a CDR10I library spanning 4
nt residues via splicing by overlap extension (SOE)(Horton et al. (1990)
Biotechniques, 8, 528-535). The human WT1-D13 CDR10I scTCR library was thus
introduced into the yeast display vector by combining the linearized pCT302 vector,
WT1—D13 CDR10I y PCR product, and competent EBY100 yeast cells. The
ant library contained about 3.1 X 106 independent clones, was judged by
plating limiting dilution aliquots of yeast after electroporation. The WT1-D13
CDR10I library were FACS sorted according to the Table 2 for binding to WT1
(RMFPNAPYL, SEQ ID NO:6)/HLA.A2/lg dimers (BD DimerX).
Table 2. Sorting Conditions
1 100 nM WT1:HLA.A2 dimer
APO—conjugated goat anti-mouse secondary antibody (1:100)
2 100 nM WT1:HLA.A2 dimer
APO—conjugated goat ouse secondary dy (1:100)
3 100 nM WT1:HLA.A2 dimer
APO—conjugated goat anti-mouse secondary antibody (1:100)
4 100 nM WT1:HLA.A2 dimer
APO—conjugated goat anti-mouse secondary antibody (1:100)
200 nM WT1:HLA.A2 dimer
APO—conjugated goat anti-mouse secondary antibody (1:100)
After five rounds of selection by FACS with WT1 (RMFPNAPYL, SEQ ID
NO:6)/HLA-A2/lg dimers, a modestly positively staining population began to emerge
(Figure 6A). Clone 3.1, isolated following the 5th sort, showed a modest
binding improvement to WT1 (RMFPNAPYL, SEQ ID HLA.A2 and was used
as a template for further affinity maturation (Figure 6B).
EXAMPLE 6
CDR3 LIBRARY CONSTRUCTION AND SELECTION OF A WT1 TCR WITH FURTHER ENHANCED
BINDING TO WT1:HLA.A2,WT1.1.1
In order to further improve the affinity of the WT1 scTCR, CDR3 libraries
were generated using the WT1-D13.1 clone isolated from the WT1-D13
CDR10I y as a template. The WT1-D131 CDR3 libraries were generated by
splicing by overlap extension (SOE) PCR, making degenerate codons spanning 5
adjacent codons in each CDR3 at a time (2 libraries in the CDR3[3 loop; 2 in the
CDR30I loop). Each WT1-D131 CDR3 library was thus introduced into the yeast
y vector by combining the linearized pCT302 vector, WT1-D131 CDR3 PCR
product (i.e. 1, CDR30I2, CDR3B1, or CDR3B2 library), and competent
EBY100 yeast cells. The four resultant libraries were pooled and the ant
ed library contained about 3.5 X 106 ndent clones as determined by
plating limiting dilution aliquots of yeast after electroporation. The WT1-D13 CDR3
combined ies were FACS sorted according to the chart in Table 3 for binding to
WT1 (RMFPNAPYL, SEQ ID NO:6)/HLA.A2/lg dimers (BD DimerX).
Table 3. Sorting Conditions
1 100 nM WT1:HLA.A2 dimer
APO-conjugated goat ouse secondary antibody (1:100)
2 100 nM WT1:HLA.A2 dimer
APO-conjugated goat anti—mouse secondary antibody )
3 10 nM WT1:HLA.A2 dimer
APO-conjugated goat ouse secondary antibody (1:100)
After three rounds of selection by FACS with WT1 (RMFPNAPYL, SEQ ID
NO:8)/HLA—A2/Ig dimers, a positively staining population began to emerge (Figure
7A). Clone WT1-D13.1.1, isolated following the 3rd sort, showed increased binding to
WT1 (RMFPNAPYL, SEQ ID NO:8)/HLA.A2 (Figure 7B).
EXAMPLE 7
BINDING ANALYSIS OF HIGH AFFINITY WT1 TCR,WT1-D13.1.1
In order to assess the binding of the WT1-D13.1.1 clone isolated from
selections of CDR3 libraries, yeast ying the WT1-D13.1 .1 were titrated with
WT1 (RMFPNAPYL, SEQ ID NO:6)/HLA.A2 dimers HLA-A2-lg) and monomers
expressed and purified from Eco/i. The WT-1/A2 dimers were assayed at 160 pM to
500 nM (Figure 8A), and the monomers were assayed at 6.4 nM to 4 uM (Figure SB).
Yeast cells were then washed and analyzed by flow cytometry. Mean fluorescence
intensity (MFI) were d for each histogram versus the concentration of the WT-
A2 x. Values were normalized using nonlinear regression analysis and
Knapp values of 25 nM and 240 nM were determined for dimmer and monomer,
respectively (Figure 8C and 8D). Thus the WT1-D13.1.1 exhibited nanomolar affinity.
EXAMPLE 8
BINDING IS OF SOLUBLE HIGH AFFINITY WT1 TCR,WT1-D13.1.1
To further Show that the WT1-D13.1 .1 sch specifically bound
WT1/HLA-A2 with high-affinity, a soluble form of WT1-D13.1 .1 sch was expressed
and refolded from E. coli inclusion bodies and biotinylated via a C-terminal BirA tag
(Aggen et al. (2011) Protein Engineering, Design, & Selection, 24, 361-72; Zhang et
al. (2007) J Exp Med, 204, 49-55). The human cell line T2 (HLA-A2+) was incubated
with 1 uM Tax, MART-1, or WT1 es and washed. Biotinylated WT1—D13.1 .1
sch was titrated on T2 cells aded without peptide e 9A), or with the
negative peptide Tax (4 nM to 1 uM) (Figure QB), null peptide MART-1 (4 nM to 1
uM) (Figure 9C), WT1 (4 nM to 1 uM) (Figure 9D). The cells were washed and
incubated with SA-PE and analyzed by flow cytometry. Only cells loaded with WT1
peptide were bound by the WT1-D13.1.1 TCR (Figure 9A-D) showing that the
soluble TCR was specific for WT1. Non-linear regression of the plot of MFI versus
TCR concentration of the WT1 ion showed that the soluble TCR exhibited a
minimum KD value of 260 nM (Figure 9E).
EXAMPLE 9
SEQUENCE ANALYSIS OF THE ISOLATED TCRS FOR IMPROVED TY AGAINST THE WT1
ANTIGEN
Sequences of the WT1-specific (P22, D13, D13.1, D13.0.1 and D13.1.1)
single-chain TCRs were determined from isolated plasmids, and are shown in Figure
1. The amino acid sequences of the VB chain of P22, D13, D13.1, D13.0.1 and
D13.1.1 are set forth in SEQ ID NOs:1, 21, 21, 3 and 3, respectively, and the amino
acid sequences of the VOI chain of P22, D13, D13.1, D13.0.1 and D13.1.1 are set
forth in SEQ ID NOs:2, 22, 4, 2, and 4, respectively. Note that D13.0.1 was
constructed by removing the CDR10I mutations from the final affinity matured scTCR
D13.1.1 (SEQ ID NO: 5). The underlined amino acid ons in Figure 1 indicate
mutations that arose from error-prone library selections for stabilizing mutations.
The amino acid positions in boxes show the ty enhancing mutations that were
selected from CDR libraries.
EXAMPLE 10
IN VITRo ACTIVITY OF THE WT1-P22, WT1-D131, WT1-D13.0.1 AND WT1-D13.1.1
TCRs IN CD8 AND CD4 T CELLS
To assess the ty of the different WT1-specific TCRs in T cells, CD8
(Figure 10A) and CD4 (Figure 108) T cells were isolated from AAD transgenic mice
(these are mice that have a hybrid class I gene ting of the (11 and (12 s
of HLA-A2 and the Q3 domain of the mouse Db; these AAD mice are available from
Jackson Laboratories). The cells were activated with beads coupled with anti-CD3
and anti-CD28 antibodies and then transduced with the WT1-P22, WT1-D13.1, WT1-
D13.0.1 and WT1—D13.1.1 TCRs (sequences did not contain the D13 stabilizing
mutations, VB F488 and D51 G), as described in Chervin et al, (2013) Gene Ther.
(6):634-44. T cells were incubated with different concentrations of the peptide
WT1 and AAD blast cells prepared by avilin A stimulations of splenic cells
from AAD mice. After a 24-hour incubation, atants were analyzed for lFN-v
concentration using an ELISA. CD8 T cells showed the greatest activity with the
D13.0.1 and D13.1.1 TCRs (Figure 10A). CD4 T cells were only activated with the
1 TCR, showing that D13.1.1 can mediate activity ndently of CD8
(Figure 108). No reactivity has been observed with other HLA-A2 g peptides
such as MART1.
EXAMPLE 11
WT1-D13.1.1 TCR IN CD8 T CELLS ARE NOT REACTIVE WITH HUMAN PEPTIDES THAT ARE
STRUCTURALLY SIMILAR TO WT1
To further determine the specificity of the high-affinity WT1-D13.1.1 TCR,
in vitro activity with peptides structurally similar to the WT1 peptide was ed. A
proteome search was conducted for peptides urally similar to the WT1 peptide,
based on conservative mutations at the 9 residues of WT1. Peptides present in the
human proteome were then accessed for g ability to HLA-A2 via prediction
algorithms. The ten peptides that were predicted to bind HLA-A2 with the highest
affinity (Figure 11) were synthesized and tested for ability to activated CD8 T cells
transduced with the high-affinity WT1-D13.1 .1 TCR. None of these peptides
displayed activity, suggesting that this TCR maintains specificity when presented
with these 10 structurally similar peptides.
EXAMPLE 12
THERAPEUTIC FORMATS OF THE WT1, WT1-D13.1 AND WT1-D13.1.1 TCRs
It is now well known that higher affinity TCRs can be used in various
formats for ing cells that express the corresponding antigen. Thus, it is clear
that the TCRs generated from the engineering strategies shown above can be used
either in soluble form or in TCR gene therapy for adoptive T cell ies, as
illustrated in Figure 12.
Materials and methods
Antibodies, peptidezHLA-A2, MACSI and Flow Cytometry Reagents
Antibodies used to detect yeast surface expression included: anti-HA
e tag (Clone HA.11; Covance), anti-hVB3 FITC dy (Clone CH92;
Beckman-Coulter), anti-hVB3.1 FITC antibody (Clone 8F10; Thermo Scientific), anti-
hVBZO antibody (Clone ELL1.4; Beckman-Coulter), anti-V02 monoclonal antibody
ted in our laboratory (data not shown), Goat-anti-mouse lgM APC (Life
logies), Goat-anti-mouse lgG F(ab’)2 AlexaFluor® 647 secondary antibody
(Invitrogen), Streptavidin-phycoerythrin (SA:PE, BD ngen), and MACS
microbeads nyl Biotec).
Peptides that bind to HLA-A2 [WT1126_134: RMFPNAPYL (SEQ ID NO:6)
were synthesized by standard F-moc fluorenyl)methoxycarbonyl) chemistry at
the Macromolecular Core Facility at Penn State sity College of Medicine
(Hershey, PA, USA). For FACS and flow cytometry analysis, recombinant soluble
dimeric HLA—A2:lg fusion protein (BDT'V' DimerX) was used. Additionally, a
monomeric HLA.A2-biotin reagent generated by the exchange of a UV-cleavable
peptide for another HLA.A2—restricted peptide in the ce of UV light was utilized
for flow cytometry and MACS selections ko et al. (2006) Nat , 1, 1120-
1132; Toebes et al. (2006) Nat Med, 12, 246-251).
Cloning and expression of sch in yeast display s
TCR variable region fragments (sch) were expressed in yeast y
plasmid pCT302 (VB-L-Vd) (Boder and Wittrup (2000) Methods Enzymol, 328, 430-
444), which contains a galactose-inducable AGA2 fusion allowing for growth in Trp
media. Induction of the sch gene involves growth of the transformed EBY100 yeast
cells to nary phase in selection media followed by transfer to galactose-
ning media. The template WT1 single-chain TCR gene was synthesized by
Genscript (Piscataway, NJ, USA) with a F498 mutation in the Vd2—domain of the
construct (Aggen et al. (2011) Protein Eng Des Sel, 24, 361-372).
The WT1—specific TCR genes were isolated from CTL clones (TCR genes
against WT1 from Phillip Greenberg; e.g. Dossett et al. (2009) Mol Ther. 17(4), 742)
and the genes were synthesized by ipt, cloned as a single-chain format (V8-
linker—Va), introduced into a yeast display vector for expression on the surface of
yeast. The schs consisted of the variable contains attached by the linker region
GSADDAKKDAAKKDGKS (SEQ ID NO:8) (Hoo et al. (1992) Proc Natl Acad Sci
USA, 89, 4759-4763; Weber et al. (2005) Proc Natl Acad Sci USA, 102, 19033—
19038; Aggen et al. (2011) Protein Eng Des Sel, 24, 361-372). The sch was
introduced into the Nhel and Xhol restrictions sites of pCT302.
Generation, display, and selection of mutated sch yeast display ies
Error-prone PCR was used to generate random mutations, as previously
described (Richman et al. (2009) Mol Immunol, 46, 902-916). CDR1 and 3 libraries
were generated using Splicing by p extension (SOE) PCR spanning 4-5
adjacent codons at a time (Horton et al. (1990) Biotechniques, 8, 528-535).
For the WT1-D13 CDR1or library, pre-SOE PCR products were generated
utilizing the following primer pairs: 5' - GGC AGC CCC ATA AAC ACA CAG TAT -3'
(Splice 4L) (SEQ ID NO:9) and 5’ - ACG ATC GCT ATA GGT GCA GTT CAA TGA
TGC AAT AGC ACC TTC CGG GAC ACT TAA TGG GCC GCT - 3’(SEQ ID NO:10),
and 5’ — ATT GCA TCA TTG AAC TGC ACC TAT AGC GAT CGT NNS NNS NNS
MTTC TTT TGG TAT AGA CAG TAC AGT GGC AAA TCC CCG — 3’ (SEQ ID
NO:11) and 5'— TAA TAC GAC TCA CTA TAG CG —3' (T7) (SEQ ID NO:12). SOE
PCR was performed with each ponding Pre-SOE along with both T7 and
Splice 4L.
For the 3.1 CDR3 libraries, pre-SOE PCR products were
generated for each of the four libraries utilizing the following primer pairs: B1: 5' -
GGC AGC CCC ATA AAC ACA CAG TAT -3' (Splice 4L) (SEQ ID NO:9). and 5’ —
TGC ACA CAG GTA CAT GGA AGT TTG ATT GGT ACT AGC GCT TTC CAG AAT
CAA ACT GAA ACG TTC TTT — 3’ (SEQ ID NO:13), and 5’ — AGT ACC AAT CAA
ACT TCC ATG TAC CTG TGT GCA NNS NNS NNS NNS NNS GAA CAG TTT TTC
GGC CCA GGT ACA AGA TTA ACG GTG — 3’ (SEQ ID NO:14) and 5'- TAA TAC
GAC TCA CTA TAG CG -3' (T7) (SEQ ID NO:12); B2: Splice 4L (SEQ ID NO:9) and
’ — TGC ACA CAG GTA CAT GGA AGT TTG ATT GGT ACT AGC GCT TTC CAG
AAT CAA ACT GAA ACG TTC TTT — 3’ (SEQ ID NO:15), and 5’ — AGT ACC AAT
CAA ACT TCC ATG TAC CTG TGT GCA AGC AGT TCC ATC NNS NNS NNS NNS
MGGC CCA GGT ACA AGA TTA ACG GTG — 3’ (SEQ ID NO:16) and T7 (SEQ
ID NO:12); o1: Splice 4L (SEQ ID NO:9) and 5’ — GGC GCA CAG GTA AGT GGC
GCT ATC TGA CGG TTG GCT ATC ACG GAT TAA CAG AGA GAC ATA CTG
GGA — 3’ (SEQ ID NO:17), and 5’ — CAA CCG TCA GAT AGC GCC ACT TAC CTG
TGC GCC NNS NNS NNS NNS NNS AAT ATG CTG ACC TTC GGT GGC GGT
ACT CGC TTA ATG — 3’ (SEQ ID NO:18) and T7 (SEQ ID ; a2: Splice 4L
(SEQ ID NO:9) and 5’ — GGC GCA CAG GTA AGT GGC GCT ATC TGA CGG TTG
GCT ATC ACG GAT TAA CAG AGA GAC ATA CTG GGA — 3’ (SEQ ID NO:19), and
’ — CAA CCG TCA GAT AGC GCC ACT TAC CTG TGC GCC GCG AAT AAC GCG
NNS NNS NNS NNS NNS TTC GGT GGC GGT ACT CGC TTA ATG — 3’ (SEQ ID
N020) and T7 (SEQ ID NO:12).
Yeast libraries were made by homologous recombination in EBY100 yeast
by electroporating error prone or SOE PCR products along with Nhel and Xhol
digested pCT302 (Horton et al. (1990) Biotechniques, 8, 528-535). The libraries were
induced in galactose-containing media (SG-CAA) for 48 h, washed with 1 mL 1%
PBS/BSA, and stained with antibodies or peptide:MHC reagents at the
concentrations indicated in Figures 4A, 5A, 6A, 8A, and 9A. Cells were washed (1
ml, 1% PBS/BSA), and the most fluorescent cells were selected using a FACS Aria
(BD ence) high-speed sorter or via MACS LS columns on a QuadroMACST'V'
tor (Miltenyl Biotec). In order to test thermal stability of isolated clones, yeast
were incubated at elevated temperature for 30 min prior to the staining protocol (data
not shown).
Isolation and staining of high ty clones
Following selections, library clones were isolated by plating limiting
dilutions. es were expanded and induced in galactose-containing media (SG-
CAA) for 48 hours, washed with 1 mL 1% PBS/BSA, and stained with various
concentrations of peptide/HLA.A2 DimerX, goat-anti-mouse lgG F(ab’)2 luor®
647 secondary antibody, or various concentrations of UV-exchanged
e/HLA.A2, SA-PE. Cells were washed (1 ml, 1% PBS/BSA) and analyzed on
an Accuri C6 flow cytometer.
Plasmids were recovered using ZymoprepTIVI Yeast Plasmid Miniprep |l
(Zymo Research) and introduced back into E. coli via heat shock transformation into
Subcloning EfficiencyTM DH5qT'V' Competent Cells (lnvitrogen). E. coli cells were
expanded and ds were isolated using QlAprep Spin Miniprep Kit (Qiagen).
Sequences of individual clones were determined by Sanger sequencing
STATEMENTS REGARDING INCORPORATION BY NCE AND
VARIATIONS
All references cited herein, for example patent nts including issued
or d patents or equivalents; patent application publications; and nonpatent
literature documents or other source al; are hereby incorporated by reference
herein in their entireties, as though individually incorporated by reference, to the
extent each nce is at least partially not inconsistent with the disclosure in this
application (for example, a reference that is lly inconsistent is incorporated by
reference except for the partially inconsistent portion of the reference).
All patents and publications mentioned in the specification are indicative of
the levels of skill of those skilled in the art to which the disclosure pertains.
References cited herein are incorporated by reference herein in their entirety to
indicate the state of the art, in some cases as of their filing date, and it is intended
that this information can be employed herein, if needed, to exclude (for example, to
disclaim) ic embodiments that are in the prior art or to use s or
materials that are in the state of the art without the specific inclusion of the methods
or materials in the disclosure herein. For example, when a compound is claimed, it
should be understood that compounds known in the prior art, ing certain
nds disclosed in the references disclosed herein cularly in nced
patent documents), are not intended to be included in the claim.
When a Markush group or other grouping is used herein, all individual
members of the group and all combinations and binations possible of the
group are intended to be individually included in the disclosure.
Where the terms ise”, “comprises”, “comprised”, or “comprising” are
used herein, they are to be interpreted as specifying the ce of the stated
features, integers, steps, or components referred to, but not to preclude the
presence or addition of one or more other feature, integer, step, component, or
group thereof. Separate embodiments of the disclosure are also intended to be
assed n the terms “comprising” or “comprise(s)” or “comprised” are
optionally replaced with the terms, analogous in grammar, e.g.;
“consisting/consist(s)” or “consisting essentially of/consist(s) essentially of” to
thereby describe further embodiments that are not necessarily nsive. For
clarification, as used herein “comprising” is synonymous with “having,” "including,"
"containing," or "characterized by," and is inclusive or open-ended and does not
exclude onal, unrecited elements or method steps. As used herein, sting
of" excludes any element, step, component, or ingredient not specified in the claim
element. As used herein, "consisting essentially of" does not exclude materials or
steps that do not materially affect the basic and novel characteristics of the claim
(e.g., not affecting an active ingredient). In each instance herein any of the terms
"comprising", "consisting essentially of" and "consisting of" may be replaced with
either of the other two terms. The disclosure illustratively described herein suitably
may be practiced in the absence of any element or ts, limitation or limitations
which is not specifically disclosed herein.
The disclosure has been described with reference to various specific and
preferred embodiments and techniques. However, it should be understood that many
variations and modifications may be made while remaining within the spirit and
scope of the sure. It will be appreciated by one of ordinary skill in the art that
compositions, methods, devices, device elements, materials, optional features,
procedures and ques other than those specifically described herein can be
applied to the practice of the disclosure as broadly disclosed herein without resort to
undue experimentation. All art-known functional equivalents of compositions,
methods, devices, device elements, als, procedures and techniques bed
herein; and ns thereof; are intended to be encompassed by this disclosure.
Whenever a range is disclosed, all subranges and individual values are intended to
be encompassed. This disclosure is not to be limited by the embodiments disclosed,
including any shown in the drawings or exemplified in the specification, which are
given by way of example or illustration and not of limitation. Some references
ed herein are orated by reference herein to e details concerning
additional starting materials, additional methods of synthesis, and additional methods
of analysis and onal uses of the disclosure.
One d in the art would readily appreciate that the present disclosure
is well adapted to carry out the objects and obtain the ends and advantages
mentioned, as well as those nt therein. The compositions and methods and
accessory methods described herein as presently representative of preferred
embodiments are exemplary and are not intended as limitations on the scope of the
disclosure. Changes therein and other uses will occur to those skilled in the art,
which are encompassed within the spirit of the disclosure.
NCES
1. Addo M. M., Draenert R., Rathod A., Verrill C. L., Davis B. T., Gandhi R. T.,
Robbins G. K., Basgoz N. 0., Stone D. R., Cohen D. E., Johnston M. N., Flynn T.,
Wurcel A. G., Rosenberg E. 8., Altfeld M. and Walker B. D. (2007) Fully
Differentiated HIV-1 Specific CD8+ T Effector Cells Are More Frequently Detectable
in Controlled than in Progressive HIV—1 Infection. PLoS ONE 2, e321.
2. Aggen D. H., Chervin A. 8., lnsaidoo F. K., Piepenbrink K., H., Baker B. M.
and Kranz D. M. (2011) Identification and engineering of human variable regions that
allow expression of stable single-chain T cell receptors. Protein ering, Design,
& Selection 24, 361-72.
3. Anikeeva N., Mareeva T., Liu W. and Sykulev Y. (2009) Can eric T-
cell or be used as a tool to detect viral peptide epitopes on infected cells? Clin
Immunol 130, 98-109.
4. Armstrong K. M., Piepenbrink K. H. and Baker B. M. (2008) Conformational
changes and flexibility in T-cell receptor recognition of peptide-MHC complexes.
Biochem J 415, .
. ld R. and Jakobsen B. K. (2006) Making ffinity T-cell receptors:
a new class of targeted therapeutics. lDrugs 9, 554-9.
6. Bargou R., Leo E., Zugmaier G., Klinger M., Goebeler M., Knop S.,
Noppeney R., Viardot A., Hess G., Schuler M., Einsele H., Brandl C., Wolf A.,
Kirchinger P., Klappers P., Schmidt M., Riethmuller G., Reinhardt C., Baeuerle P. A.
and Kufer P. (2008) Tumor regression in cancer patients by very low doses of a T
cell-engaging antibody. e 321, 974-7.
7. Benatuil L., Perez J. M., Belk J. and Hsieh C. M. (2010) An improved yeast
transformation method for the generation of very large human antibody libraries.
Protein Eng Des Sel 23, 155—9.
8. Bird R. E., Hardman K. D., Jacobson J. W., Johnson 8., Kaufman B. M.,
Lee S. M., Lee T., Pope S. H., Riordan G. S. and Whitlow M. (1988) Single-chain
antigen-binding proteins. Science 242, 423-426.
9. Boder E. T. and p K. D. (1997) Yeast surface display for screening
combinatorial polypeptide libraries. Nat. h. 15, 553-557.
. Boder E. T. and Wittrup K. D. (2000) Yeast surface display for directed
evolution of protein expression, affinity, and stability. Methods Enzymol 328, 430-44.
11. Boon T. and Old L. J. (1997) Cancer tumor ns. Curr Opin Immunol 9,
681-3.
12. Borbulevych O. Y., Santhanagopolan S. M., Hossain M. and Baker B. M.
(2011) TCRs used in cancer gene therapy cross-react with MART-1/Melan-A tumor
antigens via distinct mechanisms. J Immunol 187, 2453-63.
13. Brower V. (1997) Enbrel's phase III reinforces prospects in RA [news]. Nat
Biotechnol 15, 1240.
14. Bulek A. M., Cole D. K., a A., Dolton G., Gras 8., Madura F., Fuller
A., Miles J. J., Gostick E., Price D. A., Drijfhout J. W., Knight R. R., Huang G. C.,
Lissin N., Molloy P. E., Wooldridge L., Jakobsen B. K., Rossjohn J., Peakman M.,
Rizkallah P. J. and Sewell A. K. (2012) Structural basis for the killing of human beta
cells by CD8(+) T cells in type 1 diabetes. Nat Immunol 13, 283-9.
. Cheever M. A., Allison J. P., Ferris A. S., Finn O. J., Hastings B. M., Hecht
T. T., Mellman |., ville S. A., Viner J. L., Weiner L. M. and Matrisian L. M.
(2009) The prioritization of cancer antigens: a national cancer institute pilot project
for the acceleration of translational research. Clin Cancer Res 15, 5323-37.
16. Chervin A. S., Aggen D. H., Raseman J. M. and Kranz D. M. (2008)
Engineering higher affinity T cell receptors using a T cell display system. J Immunol
Methods 339, 175-84.
17. Chervin AS, Stone JD, Soto CM, Engels B, Schreiber H, Roy EJ, and
Kranz DM. (2013) Design of T-cell receptor libraries with diverse binding properties
to examine adoptive T-cell responses. Gene Ther. 20(6):634-44
18. Colby D. W., g B. A., Graff C. P., Yeung Y. A., Swers J. S. and
Wittrup K. D. (2004) Engineering dy affinity by yeast surface display. Methods
Enzymol 388, .
19. Davis M. M. and Bjorkman P. J. (1988) T-cell antigen receptor genes and
T-cell recognition. Nature 334, 395—402.
. Davis M. M., Boniface J. J., Reich Z., Lyons D., Hampl J., Arden B. and
Chien Y. (1998) Ligand recognition by alpha beta T cell receptors. Annu Rev
Immunol 16, 523-544.
21. Ding Y. H., Baker B. M., Garboczi D. N., Biddison W. E. and Wiley D. C.
(1999) Four A6—TCR/peptide/H LA-A2 ures that generate very different T cell
signals are nearly identical. Immunity 11, 45-56.
22. Foote J. and Eisen H. N. (2000) ng the affinity g for dies
and T cell receptors. Proc Natl Acad Sci U S A 97, 81.
23. zi D. N., Ghosh P., Utz U., Fan Q. R., Biddison W. E. and Wiley D.
C. (1996) Structure of the complex between human T-cell receptor, viral peptide and
HLA-A2. Nature 384, 134-141.
24. Garcia K. C., Adams J. J., Feng D. and Ely L. K. (2009) The molecular
basis of TCR germline bias for MHC is surprisingly simple. Nat Immunol 10, 143—7.
. Haidar J. N., Pierce B., Yu Y., Tong W., Li M. and Weng Z. (2009)
Structure-based design of a T—cell receptor leads to nearly 100-fold improvement in
binding affinity for pepMHC. Proteins 74, 948-60.
26. Harkiolaki M., Holmes S. L., Svendsen P., Gregersen J. W., Jensen L. T.,
McMahon R., Friese M. A., van Boxel G., Etzensperger R., Tzartos J. S., Kranc K.,
Sainsbury S., Harlos K., Mellins E. D., Palace J., Esiri M. M., van der Merwe P. A.,
Jones E. Y. and Fugger L. (2009) T cell-mediated autoimmune disease due to low-
affinity eactivity to common microbial peptides. Immunity 30, 348-57.
27. Hawse W. F., on M. M., Joyce M. V., Hellman L. M., Hossain M.,
Ryan V., Pierce B. G., Weng Z. and Baker B. M. (2012) Cutting edge: evidence for a
dynamically driven T cell signaling mechanism. J Immunol 188, 5819-23.
28. Holler P. D., cki L. K. and Kranz D. M. (2003) TCRs with high affinity
for foreign pMHC show self-reactivity. Nat Immunol 4, 55—62.
29. Holler P. D., Holman P. 0., Shusta E. V., O'Herrin S., p K. D. and
Kranz D. M. (2000) In vitro evolution of a T cell receptor with high affinity for
peptide/MHC. Proc Natl Acad Sci U S A 97, 5387—92.
. Holliger P., Prospero T. and Winter G. (1993) "Diabodies": small bivalent
and bispecific antibody fragments. Proc Natl Acad Sci U S A 90, 6444-8.
31. Hoogenboom H. R. (2005) Selecting and screening recombinant antibody
libraries. Nat Biotechnol 23, 1105—16.
32. Jarvis L. M. (2012) Rethinking Antibody-Drug Conjugates. Chemical and
Engineering News 90, 12-18.
33. Kessels H. W., van Den Boom M. D., Spits H., erg E. and
Schumacher T. N. (2000) Changing T cell specificity by retroviral T cell receptor
y. Proc Natl Acad Sci U S A 97, 14578—83.
34. Kieke M. C., Shusta E. V., Boder E. T., Teyton L., p K. D. and Kranz
D. M. (1999) Selection of functional T cell receptor mutants from a yeast surface-
display library. Proc Natl Acad Sci U S A 96, 5651—6.
. Lauck F., Smith C. A., Friedland G. F., Humphris E. L. and Kortemme T.
(2010) RosettaBackrub--a web server for flexible backbone protein structure
modeling and design. Nucleic Acids Res 38, W569-75.
36. Li Y., Moysey R., Molloy P. E., Vuidepot A. L., Mahon T., Baston E., Dunn
S., Liddy N., Jacob J., Jakobsen B. K. and Boulter J. M. (2005) Directed evolution of
human T-cell receptors with picomolar ties by phage display. Nat Biotechnol 23,
349-54.
37. Liddy N., Bossi 6., Adams K. J., Lissina A., Mahon T. M., Hassan N. J.,
et J., Bianchi F. C., ey N. J., Ladell K., Gostick E., Sewell A. K., Lissin
N. M., Harwood N. E., Molloy P. E., Li Y., Cameron B. J., Sami M., Baston E. E.,
Todorov P. T., Paston S. J., Dennis R. E., Harper J. V., Dunn S. M., ld R.,
Johnson A., McGrath Y., Plesa G., June C. H., Kalos M., Price D. A., Vuidepot A.,
Williams D. D., Sutton D. H. and Jakobsen B. K. (2012) Monoclonal TCR-redirected
tumor cell killing. Nat Med.
38. -Greenfeld D. and Benhar l. (2012) Risks and untoward toxicities of
antibody-based immunoconjugates. Adv Drug Deliv Rev.
39. Manning T. C. and Kranz D. M. (1999) Binding energetics of T—cell
receptors: correlation with immunological consequences. Immunology Today 20,
417-422.
40. Marrack P., Scott-Browne J. P., Dai S., Gapin L. and Kappler J. W. (2008)
Evolutionarily conserved amino acids that control TCR-MHC interaction. Annu Rev
Immunol 26, 171-203.
41. Marsh S. G. E., Parham P. and Barber L. D. (2000) The HLA Facts Book.
Academic Press, .
42. Mason D. (1998) A very high level of eactivity is an essential e
of the T- cell or. Immunol Today 19, 395-404.
43. Miller B. R., Demarest S. J., Lugovskoy A., Huang P, Wu X., Snyder W. B.,
Croner L. J., Wang N., Amatucci A., Michaelson J. S. and Glaser S. M. (2010)
Stability engineering of scFvs for the development of bispecific and multivalent
antibodies. Protein Eng Des Sel 23, 549-57.
44. Molloy P. E., Sewell A. K. and Jakobsen B. K. (2005) Soluble T cell
receptors: novel immunotherapies. Curr Opin Pharmacol 5, 438-43.
45. Murphy K. (2012) Janeway's immunobiology. Garland Science, New York.
46. Nold M. F., Nold-Petry C. A., Zepp J. A., Palmer B. E., Bufler P. and
Dinarello C. A. (2010) lL-37 is a ental inhibitor of innate immunity. Nat
Immunol 11,1014-22.
47. Pastan l., Hassan R., Fitzgerald D. J. and Kreitman R. J. (2006)
lmmunotoxin therapy of cancer. Nat Rev Cancer 6, 559-65.
48. Pierce B. G., Haidar J. N., Yu Y. and Weng Z. (2010) Combinations of
ty—enhancing mutations in a T cell receptor reveal highly nonadditive s
within and between complementarity determining regions and chains. Biochemistry
49, 7050-9.
49. Porter D. L., Levine B. L., Kalos M., Bagg A. and June C. H. (2011)
Chimeric antigen receptor—modified T cells in chronic lymphoid leukemia. N Engl J
Med 365, 725-33.
50. Reichert J. M. and Valge—Archer V. E. (2007) Development trends for
monoclonal antibody cancer therapeutics. Nat Rev Drug Discov 6, 349-56.
51. Ricart A. D. and Tolcher A. W. (2007) Technology insight: cytotoxic drug
immunoconjugates for cancer therapy. Nat Clin Pract Oncol 4, 245-55.
52. Richman S. A., Aggen D. H., Dossett M. L., eyer D. L., Allen P. M.,
Greenberg P. D. and Kranz D. M. (2009) Structural es of T cell receptor
variable regions that enhance domain stability and enable expression as single-chain
ValphaVbeta fragments. Mol Immunol 46, 902-16.
53. Richman S. A. and Kranz D. M. (2007) Display, engineering, and
applications of antigen-specific T cell receptors. Biomol Eng 24, 361-73.
54. Rock K. L. and Goldberg A. L. (1999) Degradation of cell ns and the
generation of MHC class I- presented peptides. Annu Rev Immunol 17, 739-79.
55. Rudolph M. G., Stanfield R. L. and Wilson |. A. (2006) How TCRs bind
MHCs, peptides, and ptors. Annu Rev Immunol 24, 419-66.
56. Sadelain M., Brentjens R. and Riviere l. (2009) The e and potential
pitfalls of chimeric antigen receptors. Curr Opin Immunol 21, 215-23.
57. Sami M., Rizkallah P. J., Dunn S., Molloy P., Moysey R., Vuidepot A.,
Baston E., Todorov R, Li Y., Gao F., Boulter J. M. and Jakobsen B. K. (2007)
Crystal structures of high affinity human T-cell receptors bound to e major
histocompatibility complex reveal native diagonal binding geometry. Protein Eng Des
Sel 20, 397—403.
58. Schrama D., Reisfeld R. A. and Becker J. C. (2006) Antibody targeted
drugs as cancer therapeutics. Nat Rev Drug Discov 5, .
59. Scott J. K. and Smith G. P. (1990) Searching for peptide ligands with an
epitope library. Science 249, .
60. Skowera A., Ellis R. J., Varela-Calvino R., Arif S., Huang G. C., Van-Krinks
C., a A., Rackham C., Allen J. 8., Tree T. |., Zhao M., Dayan C. M., Sewell A.
K., Unger W. W., out J. W., Ossendorp F., Roep B. O. and Peakman M. (2008)
CTLs are targeted to kill beta cells in patients with type 1 diabetes through
recognition of a glucose-regulated preproinsulin epitope. J Clin Invest 118, 3390-
402.
61. Smith C. A. and me T. (2008) Backrub-like backbone simulation
recapitulates natural protein conformational variability and improves mutant side-
chain prediction. J Mol Biol 380, 742-56.
62. 800 Hoo W. F., Lacy M. J., Denzin L. K., Voss E. W. J., n K. D. and
Kranz D. M. (1992) Characterization of a single-chain T cell receptor expressed in E.
Coli. Proc. Natl. Acad. Sci. 89, 4759-4763.
63. Starr T. K., Jameson S. C. and Hogquist K. A. (2003) Positive and negative
selection ofT cells. Annu Rev Immunol 21, 139-76.
64. Starwalt S. E., Masteller E. L., one J. A. and Kranz D. M. (2003)
Directed evolution of a single-chain class II MHC product by yeast display. Protein
Eng 16, .
65. Stone J. D., Chervin A. S., Aggen D. H. and Kranz D. M. (2012) T cell
receptor engineering. Methods Enzymol 503, 189-222.
66. Stone J. D., Yin Y., Mo M., Weber K. S., eyer D. L., Allen P. M.,
Mariuzza R. A. and Kranz D. M. (2012) Engineering High-Affinity T Cell Receptor/
Cytokine Fusions for Therapeutic Targeting. ln Protein Engineering (Edited by
Kaumaya P.). lnTech.
67. Stroncek D. F., Berger C., Cheever M. A., Childs R. W., Dudley M. E.,
Flynn P., Gattinoni L., Heath J. R., Kalos M., Marincola F. M., Miller J. S.,
Mostoslavsky 6., Powell D. J., Jr., Rao M., Restifo N. P., Rosenberg S. A., O'Shea J.
and Melief C. J. (2012) New directions in cellular therapy of cancer: a summary of
the summit on cellular therapy for cancer. J Transl Med 10, 48.
68. Swers J. S., Kellogg B. A. and p K. D. (2004) Shuffled antibody
libraries d by in vivo homologous ination and yeast surface display.
Nucleic Acids Res 32, e36.
69. Tayal V. and Kalra B. S. (2008) Cytokines and anti-cytokines as
therapeutics——an update. Eur J Pharmacol 579, 1-12.
70. Thakur A. and Lum L. G. (2010) Cancer therapy with bispecific antibodies:
Clinical experience. Curr Opin Mol Ther 12, 340-9.
71. Tonegawa S. (1988) Nobel lecture in logy or medicine--1987.
Somatic generation of immune diversity. In Vitro Cell Dev Biol 24, 253-65.
72. Tsomides T. J., Aldovini A., Johnson R. P., Walker B. D., Young R. A. and
Eisen H. N. (1994) Naturally sed viral peptides recognized by cytotoxic T
lymphocytes on cells chronically infected by human immunodeficiency virus type 1. J
Exp Med 180, 1283-93.
73. Turner D. J., Ritter M. A. and George A. J. (1997) Importance of the linker
in expression of single-chain Fv antibody fragments: optimisation of peptide
sequence using phage display technology. J Immunol Methods 205, 43-54.
74. Utz U., Banks D., Jacobson S. and Biddison W. E. (1996) Analysis of the T-
cell receptor repertoire of human T—cell leukemia virus type 1 (HTLV-1) Tax-specific
CD8+ cytotoxic T cytes from patients with HTLVassociated disease:
evidence for oligoclonal expansion. J Virol 70, .
75. Varela-Rohena A., Molloy P. E., Dunn S. M., Li Y., Suhoski M. M., Carroll
R. G., Milicic A., Mahon T., Sutton D. H., Laugel B., Moysey R., Cameron B. J.,
ot A., Purbhoo M. A., Cole D. K., Phillips R. E., June C. H., Jakobsen B. K.,
Sewell A. K. and Riley J. L. (2008) Control of HIV-1 immune escape by CD8 T cells
expressing enhanced T-cell receptor. Nat Med 14, 1390-5.
76. Weber K. S., eyer D. L., Allen P. M. and Kranz D. M. (2005) Class
II-restricted T cell receptor engineered in vitro for higher affinity retains peptide
specificity and function. Proc Natl Acad Sci U S A 102, 19033-8.
77. Wong R. L., Liu B., Zhu X., You L., Kong L., Han K. P., Lee H. |., Chavaillaz
P. A., Jin M., Wang Y., Rhode P. R. and Wong H. C. (2011) Interleukin-
:lnterleukin—15 receptor alpha scaffold for creation of multivalent targeted immune
molecules. Protein Eng Des Sel 24, 373-83.
U.S. s
7,569,357; Filed 20—Feb-04; Issued 4-Aug-09; Board of Trustees University of
Illinois. High affinity TCR proteins and methods.
7,465,787; Filed 16—Dec-03; Issued 16-Dec-08; Board of Trustees University
of Illinois. Yeast cell surface display of ns and uses therof.
6,759,243; Filed 6-Dec-00; Issued 6-Jul-04; Board of Trustees University of Illinois.
High affinity TCR proteins and methods.
6,699,658; Filed 20—Jan-98 ; Issued 2—Mar-04; Board of Trustees University of
Illinois. Yeast cell surface y of ns and uses therof.
6,696,251; Filed 28—Nov-00; Issued 24-Feb—04; Board of Trustees sity of
is. Yeast cell e display of proteins and uses therof.
6,423,538; Filed 28—Nov-00; Issued 23-Jul—02; Board of Trustees University of
Illinois. Yeast cell surface y of proteins and uses therof.
6,300,065; Filed 26-Aug—98; Issued 9-Oct—01; Board of Trustees University of
is. Yeast cell surface display of proteins and uses therof.
8,143,376; Filed 18-May—05; Issued 27-Mar-12; Immunocore Limited; High affinity
NY-ESO T cell receptor.
8,088,379; Filed -07; Issued 3-Jan-12; Immunocore Limited; Modified T cell
receptors and related materials and methods.
8,017,730; Filed -06; Issued 13-Sep-11; Immunocore Limited; T cell
receptors which bind to WGFVRACL-HLA-A24.
7,763,718; Filed 29-Oct-07; Issued 27-Jul-10; Immunocore Limited; Soluble T cell
receptors.
604; Filed 9-Jul-03; Issued 23-Feb-10; Immunocore Limited; Modified soluble
T cell receptor.
410; Filed 7-Oct—08; Issued 27-Oct-09; Immunocore Limited; Method of
improving T cell receptors.
7,569,664; Filed 3-Oct—03; Issued 4-Aug-09; Immunocore Limited; Single chain
recombinant T cell receptors.
8,105,830; Filed 5-Nov-02; Issued 31-Jan—12; Altor Bioscience Corporation;
Polyspecific binding molecules and uses therof.
6,534,633; Filed 21—Oct-99; 18-Mar-03; Altor ence Corporation; Polyspecific
binding les and uses therof.
Claims (9)
1. A ed T cell receptor, or n-binding nt thereof, comprising a V and a V derived from a wild type T cell receptor, n the V and V, respectively, comprise the amino acid sequence set forth in SEQ ID NOs:2 and 3; SEQ ID NOs:4 and 3; SEQ ID NOs:4 and 21; or SEQ ID NOs:22 and 21.
2. The modified T cell receptor of claim 1, wherein the modified T cell receptor comprises the V amino acid sequence set forth in SEQ ID NO:3 and the V amino acid sequence set forth in SEQ ID NO:4.
3. The modified T cell receptor of claim 1, wherein the modified T cell or ses a single-chain T cell receptor with the amino acid sequence set forth in SEQ ID NO:5.
4. The modified T cell receptor of claim 1, comprising the V amino acid sequence set forth in SEQ ID NO:2 and the V amino acid sequence set forth in SEQ ID NO:3.
5. The modified T cell receptor of any one of claims 1-4 that binds to a complex of the peptide WT1 and the HLA-A2 molecule with a nanomolar or higher affinity, wherein the modified T cell receptor binds the complex with a KD value less than or equal to 10-6 M.
6. The modified T cell receptor of any one of claims 1-4 that is in soluble form.
7. A therapeutic agent that targets cancer cells that express the WT1 antigen, wherein the therapeutic agent comprises the modified T cell receptor of any one of claims 1-6.
8. A therapeutic agent that targets cancer cells that express the WT1 antigen, wherein the therapeutic agent comprises an isolated human T cell that expresses the modified T cell receptor of any one of claims 1-5.
9. Use of the modified T cell receptor of any one of claims 1-6 or the therapeutic agent of claim 7 or claim 8 in the manufacture of a medicament for treating a t having a cancer that expresses the WT1 antigen. Zflwzzzz
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201361907887P | 2013-11-22 | 2013-11-22 | |
US61/907,887 | 2013-11-22 | ||
PCT/US2014/066903 WO2015077615A1 (en) | 2013-11-22 | 2014-11-21 | Engineered high-affinity human t cell receptors |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ719720A NZ719720A (en) | 2020-09-25 |
NZ719720B2 true NZ719720B2 (en) | 2021-01-06 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2019272003B2 (en) | Engineered high-affinity human T cell receptors | |
AU2018200222B2 (en) | Engineering T-cell receptors | |
NZ719720B2 (en) | Engineered human t cell receptors with high affinity for wt1 | |
NZ719707B2 (en) | Engineered human t cell receptors with high affinity for binding survivin | |
BR112016011583B1 (en) | GENETICALLY MODIFIED HIGH AFFINITY HUMAN T CELL RECEPTORS AND THEIR USES |