WO1998018820A1 - Procedes et compositions relatifs a des immunotoxines de la proteine de fusion du ricin, pour le traitement du cancer et de maladies auto-immunes - Google Patents
Procedes et compositions relatifs a des immunotoxines de la proteine de fusion du ricin, pour le traitement du cancer et de maladies auto-immunes Download PDFInfo
- Publication number
- WO1998018820A1 WO1998018820A1 PCT/US1997/019577 US9719577W WO9818820A1 WO 1998018820 A1 WO1998018820 A1 WO 1998018820A1 US 9719577 W US9719577 W US 9719577W WO 9818820 A1 WO9818820 A1 WO 9818820A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- fusion protein
- ricin
- amino acid
- chain
- ligand
- Prior art date
Links
- 108010039491 Ricin Proteins 0.000 title claims abstract description 218
- 108020001507 fusion proteins Proteins 0.000 title claims abstract description 135
- 102000037865 fusion proteins Human genes 0.000 title claims abstract description 134
- 229940051026 immunotoxin Drugs 0.000 title claims abstract description 72
- 239000002596 immunotoxin Substances 0.000 title claims abstract description 72
- 231100000608 immunotoxin Toxicity 0.000 title claims abstract description 72
- 230000002637 immunotoxin Effects 0.000 title claims abstract description 71
- 206010028980 Neoplasm Diseases 0.000 title claims abstract description 51
- 208000023275 Autoimmune disease Diseases 0.000 title claims abstract description 46
- 201000011510 cancer Diseases 0.000 title claims abstract description 46
- 238000000034 method Methods 0.000 title claims abstract description 37
- 239000000203 mixture Substances 0.000 title description 3
- 210000004027 cell Anatomy 0.000 claims abstract description 226
- 230000027455 binding Effects 0.000 claims abstract description 125
- 238000009739 binding Methods 0.000 claims abstract description 117
- 239000002523 lectin Substances 0.000 claims abstract description 70
- 229930186900 holotoxin Natural products 0.000 claims abstract description 67
- 108090001090 Lectins Proteins 0.000 claims abstract description 64
- 102000004856 Lectins Human genes 0.000 claims abstract description 64
- 239000003446 ligand Substances 0.000 claims abstract description 58
- 230000004048 modification Effects 0.000 claims abstract description 55
- 238000012986 modification Methods 0.000 claims abstract description 55
- 239000013598 vector Substances 0.000 claims abstract description 36
- 230000014509 gene expression Effects 0.000 claims abstract description 33
- 150000007523 nucleic acids Chemical class 0.000 claims abstract description 30
- 108020004707 nucleic acids Proteins 0.000 claims abstract description 28
- 102000039446 nucleic acids Human genes 0.000 claims abstract description 28
- 108010001857 Cell Surface Receptors Proteins 0.000 claims abstract description 27
- 239000003937 drug carrier Substances 0.000 claims abstract description 10
- 210000003527 eukaryotic cell Anatomy 0.000 claims abstract description 8
- 102000006240 membrane receptors Human genes 0.000 claims abstract 6
- 238000006467 substitution reaction Methods 0.000 claims description 51
- 239000003053 toxin Substances 0.000 claims description 39
- 241000699670 Mus sp. Species 0.000 claims description 38
- 231100000765 toxin Toxicity 0.000 claims description 38
- 108700012359 toxins Proteins 0.000 claims description 38
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 33
- 235000000346 sugar Nutrition 0.000 claims description 25
- 108010002350 Interleukin-2 Proteins 0.000 claims description 23
- 230000001988 toxicity Effects 0.000 claims description 23
- 231100000419 toxicity Toxicity 0.000 claims description 23
- 150000001413 amino acids Chemical class 0.000 claims description 22
- 125000003118 aryl group Chemical group 0.000 claims description 21
- 102000010789 Interleukin-2 Receptors Human genes 0.000 claims description 18
- 108010038453 Interleukin-2 Receptors Proteins 0.000 claims description 18
- 230000009467 reduction Effects 0.000 claims description 18
- 241000282414 Homo sapiens Species 0.000 claims description 13
- 208000032839 leukemia Diseases 0.000 claims description 9
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 claims description 6
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 claims description 6
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 claims description 6
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 6
- 210000003714 granulocyte Anatomy 0.000 claims description 6
- 101800003838 Epidermal growth factor Proteins 0.000 claims description 5
- 229940116977 epidermal growth factor Drugs 0.000 claims description 5
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 claims description 5
- 206010025323 Lymphomas Diseases 0.000 claims description 3
- 208000031261 Acute myeloid leukaemia Diseases 0.000 claims description 2
- 208000009329 Graft vs Host Disease Diseases 0.000 claims description 2
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 claims description 2
- 206010029260 Neuroblastoma Diseases 0.000 claims description 2
- 208000024908 graft versus host disease Diseases 0.000 claims description 2
- 201000001441 melanoma Diseases 0.000 claims description 2
- 102100020873 Interleukin-2 Human genes 0.000 claims 3
- 102000009024 Epidermal Growth Factor Human genes 0.000 claims 2
- 241000196324 Embryophyta Species 0.000 description 93
- 102220401810 c.110G>C Human genes 0.000 description 57
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-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 48
- 108090000623 proteins and genes Proteins 0.000 description 43
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 41
- 239000008101 lactose Substances 0.000 description 41
- 102000004169 proteins and genes Human genes 0.000 description 39
- 235000018102 proteins Nutrition 0.000 description 36
- 235000001014 amino acid Nutrition 0.000 description 32
- 102220055260 rs140446520 Human genes 0.000 description 28
- 241001465754 Metazoa Species 0.000 description 25
- 241000699666 Mus <mouse, genus> Species 0.000 description 25
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 24
- 230000004927 fusion Effects 0.000 description 24
- 239000002953 phosphate buffered saline Substances 0.000 description 23
- 239000006228 supernatant Substances 0.000 description 22
- 102000000844 Cell Surface Receptors Human genes 0.000 description 21
- 208000026935 allergic disease Diseases 0.000 description 21
- 108010044715 asialofetuin Proteins 0.000 description 21
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 21
- 102000000588 Interleukin-2 Human genes 0.000 description 20
- 239000000833 heterodimer Substances 0.000 description 19
- 238000002965 ELISA Methods 0.000 description 18
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 18
- 230000003013 cytotoxicity Effects 0.000 description 18
- 231100000135 cytotoxicity Toxicity 0.000 description 18
- 231100000636 lethal dose Toxicity 0.000 description 18
- 102000005962 receptors Human genes 0.000 description 18
- 108020003175 receptors Proteins 0.000 description 18
- 238000003556 assay Methods 0.000 description 17
- 231100000566 intoxication Toxicity 0.000 description 17
- 230000035987 intoxication Effects 0.000 description 17
- 108010031099 Mannose Receptor Proteins 0.000 description 16
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N Sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 16
- 239000000427 antigen Substances 0.000 description 16
- 108091007433 antigens Proteins 0.000 description 16
- 102000036639 antigens Human genes 0.000 description 16
- 239000002609 medium Substances 0.000 description 16
- 108020004414 DNA Proteins 0.000 description 15
- 241000238631 Hexapoda Species 0.000 description 15
- 230000002950 deficient Effects 0.000 description 15
- 229940098773 bovine serum albumin Drugs 0.000 description 14
- 208000024891 symptom Diseases 0.000 description 13
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 12
- 229920000057 Mannan Polymers 0.000 description 12
- 239000012634 fragment Substances 0.000 description 12
- 241000283973 Oryctolagus cuniculus Species 0.000 description 11
- 230000000694 effects Effects 0.000 description 11
- 230000006058 immune tolerance Effects 0.000 description 11
- 239000007924 injection Substances 0.000 description 11
- 238000002347 injection Methods 0.000 description 11
- 210000003024 peritoneal macrophage Anatomy 0.000 description 11
- 238000010790 dilution Methods 0.000 description 10
- 239000012895 dilution Substances 0.000 description 10
- 230000003834 intracellular effect Effects 0.000 description 10
- 238000000746 purification Methods 0.000 description 10
- 230000014616 translation Effects 0.000 description 10
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 9
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 9
- 239000012894 fetal calf serum Substances 0.000 description 9
- 230000006870 function Effects 0.000 description 9
- 239000000499 gel Substances 0.000 description 9
- 231100000568 intoxicate Toxicity 0.000 description 9
- 108090000765 processed proteins & peptides Proteins 0.000 description 9
- 239000011780 sodium chloride Substances 0.000 description 9
- 238000011282 treatment Methods 0.000 description 9
- 241000701447 unidentified baculovirus Species 0.000 description 9
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 8
- 230000034994 death Effects 0.000 description 8
- 231100000517 death Toxicity 0.000 description 8
- 238000002474 experimental method Methods 0.000 description 8
- 229930182830 galactose Natural products 0.000 description 8
- 230000028993 immune response Effects 0.000 description 8
- 238000003119 immunoblot Methods 0.000 description 8
- 210000004962 mammalian cell Anatomy 0.000 description 8
- 238000002360 preparation method Methods 0.000 description 8
- 238000001243 protein synthesis Methods 0.000 description 8
- 238000010276 construction Methods 0.000 description 7
- 238000000338 in vitro Methods 0.000 description 7
- 230000006698 induction Effects 0.000 description 7
- 230000002147 killing effect Effects 0.000 description 7
- 238000009533 lab test Methods 0.000 description 7
- 230000001404 mediated effect Effects 0.000 description 7
- 102000004196 processed proteins & peptides Human genes 0.000 description 7
- 230000002829 reductive effect Effects 0.000 description 7
- 229930195727 α-lactose Natural products 0.000 description 7
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 6
- 229920001213 Polysorbate 20 Polymers 0.000 description 6
- 101000762949 Pseudomonas aeruginosa (strain ATCC 15692 / DSM 22644 / CIP 104116 / JCM 14847 / LMG 12228 / 1C / PRS 101 / PAO1) Exotoxin A Proteins 0.000 description 6
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 6
- YJQCOFNZVFGCAF-UHFFFAOYSA-N Tunicamycin II Natural products O1C(CC(O)C2C(C(O)C(O2)N2C(NC(=O)C=C2)=O)O)C(O)C(O)C(NC(=O)C=CCCCCCCCCC(C)C)C1OC1OC(CO)C(O)C(O)C1NC(C)=O YJQCOFNZVFGCAF-UHFFFAOYSA-N 0.000 description 6
- 238000007792 addition Methods 0.000 description 6
- 238000001514 detection method Methods 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 201000010099 disease Diseases 0.000 description 6
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 6
- 239000013604 expression vector Substances 0.000 description 6
- 150000008195 galaktosides Chemical class 0.000 description 6
- 230000001900 immune effect Effects 0.000 description 6
- 238000010166 immunofluorescence Methods 0.000 description 6
- 239000011159 matrix material Substances 0.000 description 6
- 239000013612 plasmid Substances 0.000 description 6
- 229920002401 polyacrylamide Polymers 0.000 description 6
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 6
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 6
- 229920001184 polypeptide Polymers 0.000 description 6
- 210000001519 tissue Anatomy 0.000 description 6
- 230000009261 transgenic effect Effects 0.000 description 6
- 210000004881 tumor cell Anatomy 0.000 description 6
- ZHSGGJXRNHWHRS-VIDYELAYSA-N tunicamycin Chemical compound O([C@H]1[C@@H]([C@H]([C@@H](O)[C@@H](CC(O)[C@@H]2[C@H]([C@@H](O)[C@@H](O2)N2C(NC(=O)C=C2)=O)O)O1)O)NC(=O)/C=C/CC(C)C)[C@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1NC(C)=O ZHSGGJXRNHWHRS-VIDYELAYSA-N 0.000 description 6
- MEYZYGMYMLNUHJ-UHFFFAOYSA-N tunicamycin Natural products CC(C)CCCCCCCCCC=CC(=O)NC1C(O)C(O)C(CC(O)C2OC(C(O)C2O)N3C=CC(=O)NC3=O)OC1OC4OC(CO)C(O)C(O)C4NC(=O)C MEYZYGMYMLNUHJ-UHFFFAOYSA-N 0.000 description 6
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 5
- 102000003886 Glycoproteins Human genes 0.000 description 5
- 108090000288 Glycoproteins Proteins 0.000 description 5
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 5
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 5
- 239000012980 RPMI-1640 medium Substances 0.000 description 5
- 240000000528 Ricinus communis Species 0.000 description 5
- 235000004443 Ricinus communis Nutrition 0.000 description 5
- 239000007983 Tris buffer Substances 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 238000000326 densiometry Methods 0.000 description 5
- 238000011534 incubation Methods 0.000 description 5
- 208000015181 infectious disease Diseases 0.000 description 5
- 230000003993 interaction Effects 0.000 description 5
- 238000005259 measurement Methods 0.000 description 5
- 239000008188 pellet Substances 0.000 description 5
- 230000002265 prevention Effects 0.000 description 5
- 230000035945 sensitivity Effects 0.000 description 5
- 239000001488 sodium phosphate Substances 0.000 description 5
- 229910000162 sodium phosphate Inorganic materials 0.000 description 5
- 208000011580 syndromic disease Diseases 0.000 description 5
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 5
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 5
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 4
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 4
- 241000283707 Capra Species 0.000 description 4
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 4
- 238000002835 absorbance Methods 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 230000001580 bacterial effect Effects 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 231100000433 cytotoxic Toxicity 0.000 description 4
- 230000001472 cytotoxic effect Effects 0.000 description 4
- 238000002784 cytotoxicity assay Methods 0.000 description 4
- 231100000263 cytotoxicity test Toxicity 0.000 description 4
- 238000012217 deletion Methods 0.000 description 4
- 230000037430 deletion Effects 0.000 description 4
- 238000000502 dialysis Methods 0.000 description 4
- 239000000284 extract Substances 0.000 description 4
- 229910052739 hydrogen Inorganic materials 0.000 description 4
- 239000001257 hydrogen Substances 0.000 description 4
- 238000001727 in vivo Methods 0.000 description 4
- 231100000567 intoxicating Toxicity 0.000 description 4
- 230000002673 intoxicating effect Effects 0.000 description 4
- 230000000670 limiting effect Effects 0.000 description 4
- -1 lytiacase Proteins 0.000 description 4
- 210000002540 macrophage Anatomy 0.000 description 4
- 231100000252 nontoxic Toxicity 0.000 description 4
- 230000003000 nontoxic effect Effects 0.000 description 4
- 230000003287 optical effect Effects 0.000 description 4
- 230000008488 polyadenylation Effects 0.000 description 4
- 230000009257 reactivity Effects 0.000 description 4
- 230000010076 replication Effects 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- 231100000331 toxic Toxicity 0.000 description 4
- 230000002588 toxic effect Effects 0.000 description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 3
- 102400001368 Epidermal growth factor Human genes 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 108700007698 Genetic Terminator Regions Proteins 0.000 description 3
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 3
- 102220487557 Phospholipase A and acyltransferase 1_K40M_mutation Human genes 0.000 description 3
- 239000002202 Polyethylene glycol Substances 0.000 description 3
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 3
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 3
- 241000256251 Spodoptera frugiperda Species 0.000 description 3
- 230000006907 apoptotic process Effects 0.000 description 3
- 230000001363 autoimmune Effects 0.000 description 3
- 239000012472 biological sample Substances 0.000 description 3
- 238000001574 biopsy Methods 0.000 description 3
- 230000000903 blocking effect Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 210000001124 body fluid Anatomy 0.000 description 3
- 239000010839 body fluid Substances 0.000 description 3
- AIYUHDOJVYHVIT-UHFFFAOYSA-M caesium chloride Chemical compound [Cl-].[Cs+] AIYUHDOJVYHVIT-UHFFFAOYSA-M 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- 238000012512 characterization method Methods 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- 210000000172 cytosol Anatomy 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 239000003814 drug Substances 0.000 description 3
- 230000008030 elimination Effects 0.000 description 3
- 238000003379 elimination reaction Methods 0.000 description 3
- 210000002950 fibroblast Anatomy 0.000 description 3
- 230000013595 glycosylation Effects 0.000 description 3
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 3
- 238000010191 image analysis Methods 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 230000005764 inhibitory process Effects 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 201000006417 multiple sclerosis Diseases 0.000 description 3
- 238000002703 mutagenesis Methods 0.000 description 3
- 231100000350 mutagenesis Toxicity 0.000 description 3
- 230000035772 mutation Effects 0.000 description 3
- 229920001542 oligosaccharide Polymers 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 238000002559 palpation Methods 0.000 description 3
- 230000036961 partial effect Effects 0.000 description 3
- 229920001223 polyethylene glycol Polymers 0.000 description 3
- 108091033319 polynucleotide Proteins 0.000 description 3
- 102000040430 polynucleotide Human genes 0.000 description 3
- 239000002157 polynucleotide Substances 0.000 description 3
- 230000003389 potentiating effect Effects 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 238000002731 protein assay Methods 0.000 description 3
- 210000001938 protoplast Anatomy 0.000 description 3
- 239000000377 silicon dioxide Substances 0.000 description 3
- 206010041823 squamous cell carcinoma Diseases 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- 238000013518 transcription Methods 0.000 description 3
- 230000035897 transcription Effects 0.000 description 3
- 238000012546 transfer Methods 0.000 description 3
- ILWRPSCZWQJDMK-UHFFFAOYSA-N triethylazanium;chloride Chemical compound Cl.CCN(CC)CC ILWRPSCZWQJDMK-UHFFFAOYSA-N 0.000 description 3
- 230000002792 vascular Effects 0.000 description 3
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- IEJPPSMHUUQABK-UHFFFAOYSA-N 2,4-diphenyl-4h-1,3-oxazol-5-one Chemical group O=C1OC(C=2C=CC=CC=2)=NC1C1=CC=CC=C1 IEJPPSMHUUQABK-UHFFFAOYSA-N 0.000 description 2
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 2
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 2
- XZKIHKMTEMTJQX-UHFFFAOYSA-N 4-Nitrophenyl Phosphate Chemical compound OP(O)(=O)OC1=CC=C([N+]([O-])=O)C=C1 XZKIHKMTEMTJQX-UHFFFAOYSA-N 0.000 description 2
- 241000251468 Actinopterygii Species 0.000 description 2
- 231100000699 Bacterial toxin Toxicity 0.000 description 2
- 102000004127 Cytokines Human genes 0.000 description 2
- 108090000695 Cytokines Proteins 0.000 description 2
- 108010053187 Diphtheria Toxin Proteins 0.000 description 2
- 102000016607 Diphtheria Toxin Human genes 0.000 description 2
- 208000000059 Dyspnea Diseases 0.000 description 2
- 206010013975 Dyspnoeas Diseases 0.000 description 2
- 239000007995 HEPES buffer Substances 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- 239000012981 Hank's balanced salt solution Substances 0.000 description 2
- 208000002250 Hematologic Neoplasms Diseases 0.000 description 2
- 101001076292 Homo sapiens Insulin-like growth factor II Proteins 0.000 description 2
- 206010020751 Hypersensitivity Diseases 0.000 description 2
- 208000003623 Hypoalbuminemia Diseases 0.000 description 2
- 102100025947 Insulin-like growth factor II Human genes 0.000 description 2
- 231100000111 LD50 Toxicity 0.000 description 2
- 108010007013 Melanocyte-Stimulating Hormones Proteins 0.000 description 2
- 102000012750 Membrane Glycoproteins Human genes 0.000 description 2
- 108010090054 Membrane Glycoproteins Proteins 0.000 description 2
- 244000061176 Nicotiana tabacum Species 0.000 description 2
- 235000002637 Nicotiana tabacum Nutrition 0.000 description 2
- 239000000020 Nitrocellulose Substances 0.000 description 2
- 238000000636 Northern blotting Methods 0.000 description 2
- 102000000447 Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase Human genes 0.000 description 2
- 102100027467 Pro-opiomelanocortin Human genes 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 108090000829 Ribosome Inactivating Proteins Proteins 0.000 description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 2
- 102100022831 Somatoliberin Human genes 0.000 description 2
- 101710142969 Somatoliberin Proteins 0.000 description 2
- 108090000631 Trypsin Proteins 0.000 description 2
- 102000004142 Trypsin Human genes 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 208000021017 Weight Gain Diseases 0.000 description 2
- 241000269368 Xenopus laevis Species 0.000 description 2
- 230000005856 abnormality Effects 0.000 description 2
- 208000006673 asthma Diseases 0.000 description 2
- 239000000688 bacterial toxin Substances 0.000 description 2
- 238000006664 bond formation reaction Methods 0.000 description 2
- 210000004556 brain Anatomy 0.000 description 2
- 150000001720 carbohydrates Chemical group 0.000 description 2
- 229910052799 carbon Inorganic materials 0.000 description 2
- 230000030833 cell death Effects 0.000 description 2
- 239000006285 cell suspension Substances 0.000 description 2
- 230000007541 cellular toxicity Effects 0.000 description 2
- 210000003169 central nervous system Anatomy 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- NKLPQNGYXWVELD-UHFFFAOYSA-M coomassie brilliant blue Chemical compound [Na+].C1=CC(OCC)=CC=C1NC1=CC=C(C(=C2C=CC(C=C2)=[N+](CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=2C=CC(=CC=2)N(CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=C1 NKLPQNGYXWVELD-UHFFFAOYSA-M 0.000 description 2
- 230000009260 cross reactivity Effects 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- ZBCBWPMODOFKDW-UHFFFAOYSA-N diethanolamine Chemical compound OCCNCCO ZBCBWPMODOFKDW-UHFFFAOYSA-N 0.000 description 2
- 230000009266 disease activity Effects 0.000 description 2
- 238000010828 elution Methods 0.000 description 2
- 230000003511 endothelial effect Effects 0.000 description 2
- 239000003623 enhancer Substances 0.000 description 2
- 239000003365 glass fiber Substances 0.000 description 2
- 238000006206 glycosylation reaction Methods 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 230000003118 histopathologic effect Effects 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 230000001506 immunosuppresive effect Effects 0.000 description 2
- 230000002779 inactivation Effects 0.000 description 2
- 238000001802 infusion Methods 0.000 description 2
- 230000010189 intracellular transport Effects 0.000 description 2
- 231100001231 less toxic Toxicity 0.000 description 2
- KWGKDLIKAYFUFQ-UHFFFAOYSA-M lithium chloride Chemical compound [Li+].[Cl-] KWGKDLIKAYFUFQ-UHFFFAOYSA-M 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 238000002483 medication Methods 0.000 description 2
- CWWARWOPSKGELM-SARDKLJWSA-N methyl (2s)-2-[[(2s)-2-[[2-[[(2s)-2-[[(2s)-2-[[(2s)-5-amino-2-[[(2s)-5-amino-2-[[(2s)-1-[(2s)-6-amino-2-[[(2s)-1-[(2s)-2-amino-5-(diaminomethylideneamino)pentanoyl]pyrrolidine-2-carbonyl]amino]hexanoyl]pyrrolidine-2-carbonyl]amino]-5-oxopentanoyl]amino]-5 Chemical compound C([C@@H](C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(=O)OC)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CCCCN)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CCCN=C(N)N)C1=CC=CC=C1 CWWARWOPSKGELM-SARDKLJWSA-N 0.000 description 2
- 235000013336 milk Nutrition 0.000 description 2
- 239000008267 milk Substances 0.000 description 2
- 210000004080 milk Anatomy 0.000 description 2
- 231100000324 minimal toxicity Toxicity 0.000 description 2
- 238000012544 monitoring process Methods 0.000 description 2
- 229940126619 mouse monoclonal antibody Drugs 0.000 description 2
- ZIUHHBKFKCYYJD-UHFFFAOYSA-N n,n'-methylenebisacrylamide Chemical compound C=CC(=O)NCNC(=O)C=C ZIUHHBKFKCYYJD-UHFFFAOYSA-N 0.000 description 2
- 229920001220 nitrocellulos Polymers 0.000 description 2
- 150000002482 oligosaccharides Chemical class 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 208000037821 progressive disease Diseases 0.000 description 2
- 238000001742 protein purification Methods 0.000 description 2
- 239000001397 quillaja saponaria molina bark Substances 0.000 description 2
- 239000011541 reaction mixture Substances 0.000 description 2
- 230000000717 retained effect Effects 0.000 description 2
- PYWVYCXTNDRMGF-UHFFFAOYSA-N rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 2
- 238000003118 sandwich ELISA Methods 0.000 description 2
- 229930182490 saponin Natural products 0.000 description 2
- 150000007949 saponins Chemical class 0.000 description 2
- 238000002741 site-directed mutagenesis Methods 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 210000001541 thymus gland Anatomy 0.000 description 2
- 231100000816 toxic dose Toxicity 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 230000001131 transforming effect Effects 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 230000005945 translocation Effects 0.000 description 2
- 230000032258 transport Effects 0.000 description 2
- LWIHDJKSTIGBAC-UHFFFAOYSA-K tripotassium phosphate Chemical compound [K+].[K+].[K+].[O-]P([O-])([O-])=O LWIHDJKSTIGBAC-UHFFFAOYSA-K 0.000 description 2
- 239000012588 trypsin Substances 0.000 description 2
- 230000004584 weight gain Effects 0.000 description 2
- 235000019786 weight gain Nutrition 0.000 description 2
- 238000001262 western blot Methods 0.000 description 2
- 210000005253 yeast cell Anatomy 0.000 description 2
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- RDEIXVOBVLKYNT-VQBXQJRRSA-N (2r,3r,4r,5r)-2-[(1s,2s,3r,4s,6r)-4,6-diamino-3-[(2r,3r,6s)-3-amino-6-(1-aminoethyl)oxan-2-yl]oxy-2-hydroxycyclohexyl]oxy-5-methyl-4-(methylamino)oxane-3,5-diol;(2r,3r,4r,5r)-2-[(1s,2s,3r,4s,6r)-4,6-diamino-3-[(2r,3r,6s)-3-amino-6-(aminomethyl)oxan-2-yl]o Chemical compound OS(O)(=O)=O.O1C[C@@](O)(C)[C@H](NC)[C@@H](O)[C@H]1O[C@@H]1[C@@H](O)[C@H](O[C@@H]2[C@@H](CC[C@@H](CN)O2)N)[C@@H](N)C[C@H]1N.O1C[C@@](O)(C)[C@H](NC)[C@@H](O)[C@H]1O[C@@H]1[C@@H](O)[C@H](O[C@@H]2[C@@H](CC[C@H](O2)C(C)N)N)[C@@H](N)C[C@H]1N.O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N RDEIXVOBVLKYNT-VQBXQJRRSA-N 0.000 description 1
- BFSVOASYOCHEOV-UHFFFAOYSA-N 2-diethylaminoethanol Chemical compound CCN(CC)CCO BFSVOASYOCHEOV-UHFFFAOYSA-N 0.000 description 1
- OSJPPGNTCRNQQC-UWTATZPHSA-N 3-phospho-D-glyceric acid Chemical compound OC(=O)[C@H](O)COP(O)(O)=O OSJPPGNTCRNQQC-UWTATZPHSA-N 0.000 description 1
- HIQIXEFWDLTDED-UHFFFAOYSA-N 4-hydroxy-1-piperidin-4-ylpyrrolidin-2-one Chemical compound O=C1CC(O)CN1C1CCNCC1 HIQIXEFWDLTDED-UHFFFAOYSA-N 0.000 description 1
- WYWHKKSPHMUBEB-UHFFFAOYSA-N 6-Mercaptoguanine Natural products N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 1
- 108010066676 Abrin Proteins 0.000 description 1
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 1
- 229930024421 Adenine Natural products 0.000 description 1
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 1
- 208000009746 Adult T-Cell Leukemia-Lymphoma Diseases 0.000 description 1
- 208000016683 Adult T-cell leukemia/lymphoma Diseases 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- 229920000936 Agarose Polymers 0.000 description 1
- 208000035285 Allergic Seasonal Rhinitis Diseases 0.000 description 1
- 206010002556 Ankylosing Spondylitis Diseases 0.000 description 1
- 108010039627 Aprotinin Proteins 0.000 description 1
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 1
- 241000255789 Bombyx mori Species 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 241000701822 Bovine papillomavirus Species 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 206010051290 Central nervous system lesion Diseases 0.000 description 1
- 108010049048 Cholera Toxin Proteins 0.000 description 1
- 102000009016 Cholera Toxin Human genes 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 101800004637 Communis Proteins 0.000 description 1
- 206010011224 Cough Diseases 0.000 description 1
- 241000699802 Cricetulus griseus Species 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- 239000004971 Cross linker Substances 0.000 description 1
- 241000256113 Culicidae Species 0.000 description 1
- NBSCHQHZLSJFNQ-QTVWNMPRSA-N D-Mannose-6-phosphate Chemical compound OC1O[C@H](COP(O)(O)=O)[C@@H](O)[C@H](O)[C@@H]1O NBSCHQHZLSJFNQ-QTVWNMPRSA-N 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 240000006497 Dianthus caryophyllus Species 0.000 description 1
- 235000009355 Dianthus caryophyllus Nutrition 0.000 description 1
- 108010049959 Discoidins Proteins 0.000 description 1
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- LVGKNOAMLMIIKO-UHFFFAOYSA-N Elaidinsaeure-aethylester Natural products CCCCCCCCC=CCCCCCCCC(=O)OCC LVGKNOAMLMIIKO-UHFFFAOYSA-N 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 1
- 235000014066 European mistletoe Nutrition 0.000 description 1
- 108010074860 Factor Xa Proteins 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 229930182566 Gentamicin Natural products 0.000 description 1
- CEAZRRDELHUEMR-URQXQFDESA-N Gentamicin Chemical compound O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N CEAZRRDELHUEMR-URQXQFDESA-N 0.000 description 1
- 108010033128 Glucan Endo-1,3-beta-D-Glucosidase Proteins 0.000 description 1
- 244000068988 Glycine max Species 0.000 description 1
- 235000010469 Glycine max Nutrition 0.000 description 1
- 206010072579 Granulomatosis with polyangiitis Diseases 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 1
- 108010009202 Growth Factor Receptors Proteins 0.000 description 1
- 102000009465 Growth Factor Receptors Human genes 0.000 description 1
- 208000030836 Hashimoto thyroiditis Diseases 0.000 description 1
- 101710154606 Hemagglutinin Proteins 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 239000012998 Hepes-buffered hanks’ balanced salt solution Substances 0.000 description 1
- 208000017604 Hodgkin disease Diseases 0.000 description 1
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- 108010031792 IGF Type 2 Receptor Proteins 0.000 description 1
- 206010021245 Idiopathic thrombocytopenic purpura Diseases 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 1
- 235000000177 Indigofera tinctoria Nutrition 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 206010059176 Juvenile idiopathic arthritis Diseases 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- 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 1
- 101710128836 Large T antigen Proteins 0.000 description 1
- GDBQQVLCIARPGH-UHFFFAOYSA-N Leupeptin Natural products CC(C)CC(NC(C)=O)C(=O)NC(CC(C)C)C(=O)NC(C=O)CCCN=C(N)N GDBQQVLCIARPGH-UHFFFAOYSA-N 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 102000019218 Mannose-6-phosphate receptors Human genes 0.000 description 1
- 108010087870 Mannose-Binding Lectin Proteins 0.000 description 1
- 102000009112 Mannose-Binding Lectin Human genes 0.000 description 1
- 102100024930 Melatonin receptor type 1A Human genes 0.000 description 1
- 101710098568 Melatonin receptor type 1A Proteins 0.000 description 1
- 206010048723 Multiple-drug resistance Diseases 0.000 description 1
- 206010028372 Muscular weakness Diseases 0.000 description 1
- 241001477931 Mythimna unipuncta Species 0.000 description 1
- 229930182474 N-glycoside Natural products 0.000 description 1
- 241001460678 Napo <wasp> Species 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- 206010030113 Oedema Diseases 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 102000043276 Oncogene Human genes 0.000 description 1
- 101710093908 Outer capsid protein VP4 Proteins 0.000 description 1
- 101710135467 Outer capsid protein sigma-1 Proteins 0.000 description 1
- 206010034133 Pathogen resistance Diseases 0.000 description 1
- 206010034277 Pemphigoid Diseases 0.000 description 1
- 241000721454 Pemphigus Species 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 208000031845 Pernicious anaemia Diseases 0.000 description 1
- 108010081690 Pertussis Toxin Proteins 0.000 description 1
- 102000011755 Phosphoglycerate Kinase Human genes 0.000 description 1
- 102000045595 Phosphoprotein Phosphatases Human genes 0.000 description 1
- 108700019535 Phosphoprotein Phosphatases Proteins 0.000 description 1
- 102000004160 Phosphoric Monoester Hydrolases Human genes 0.000 description 1
- 108090000608 Phosphoric Monoester Hydrolases Proteins 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 108010020221 Phytolacca americana lectin B Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 208000025237 Polyendocrinopathy Diseases 0.000 description 1
- 108010039918 Polylysine Proteins 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 101710176177 Protein A56 Proteins 0.000 description 1
- 201000004681 Psoriasis Diseases 0.000 description 1
- 108020005067 RNA Splice Sites Proteins 0.000 description 1
- 206010061481 Renal injury Diseases 0.000 description 1
- 208000037656 Respiratory Sounds Diseases 0.000 description 1
- 244000152640 Rhipsalis cassutha Species 0.000 description 1
- 235000012300 Rhipsalis cassutha Nutrition 0.000 description 1
- 101710158076 Ribosome-inactivating protein Proteins 0.000 description 1
- 241000630486 Robertus Species 0.000 description 1
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- 208000032023 Signs and Symptoms Diseases 0.000 description 1
- XUIMIQQOPSSXEZ-UHFFFAOYSA-N Silicon Chemical compound [Si] XUIMIQQOPSSXEZ-UHFFFAOYSA-N 0.000 description 1
- 208000021386 Sjogren Syndrome Diseases 0.000 description 1
- 238000002105 Southern blotting Methods 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 208000031673 T-Cell Cutaneous Lymphoma Diseases 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- 101001099217 Thermotoga maritima (strain ATCC 43589 / DSM 3109 / JCM 10099 / NBRC 100826 / MSB8) Triosephosphate isomerase Proteins 0.000 description 1
- 208000031981 Thrombocytopenic Idiopathic Purpura Diseases 0.000 description 1
- 231100000777 Toxicophore Toxicity 0.000 description 1
- 201000006704 Ulcerative Colitis Diseases 0.000 description 1
- 206010047924 Wheezing Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 229960000643 adenine Drugs 0.000 description 1
- 230000001464 adherent effect Effects 0.000 description 1
- 201000006966 adult T-cell leukemia Diseases 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- 230000007815 allergy Effects 0.000 description 1
- 229960000723 ampicillin Drugs 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
- 210000004102 animal cell Anatomy 0.000 description 1
- 230000001640 apoptogenic effect Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 229960004405 aprotinin Drugs 0.000 description 1
- 210000003567 ascitic fluid Anatomy 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 201000005000 autoimmune gastritis Diseases 0.000 description 1
- 208000006424 autoimmune oophoritis Diseases 0.000 description 1
- 201000003710 autoimmune thrombocytopenic purpura Diseases 0.000 description 1
- 201000004982 autoimmune uveitis Diseases 0.000 description 1
- 238000011888 autopsy Methods 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- 108010058966 bacteriophage T7 induced DNA polymerase Proteins 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 238000002306 biochemical method Methods 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 230000008499 blood brain barrier function Effects 0.000 description 1
- 238000004820 blood count Methods 0.000 description 1
- 210000001218 blood-brain barrier Anatomy 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 108010006025 bovine growth hormone Proteins 0.000 description 1
- 229940124630 bronchodilator Drugs 0.000 description 1
- 239000000168 bronchodilator agent Substances 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 208000000594 bullous pemphigoid Diseases 0.000 description 1
- 229910052792 caesium Inorganic materials 0.000 description 1
- TVFDJXOCXUVLDH-UHFFFAOYSA-N caesium atom Chemical compound [Cs] TVFDJXOCXUVLDH-UHFFFAOYSA-N 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 244000309466 calf Species 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000015861 cell surface binding Effects 0.000 description 1
- 210000002421 cell wall Anatomy 0.000 description 1
- 239000013611 chromosomal DNA Substances 0.000 description 1
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 description 1
- 238000001142 circular dichroism spectrum Methods 0.000 description 1
- 238000002288 cocrystallisation Methods 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 230000001186 cumulative effect Effects 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 230000007423 decrease Effects 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 238000004925 denaturation Methods 0.000 description 1
- 230000036425 denaturation Effects 0.000 description 1
- 238000000432 density-gradient centrifugation Methods 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 210000001198 duodenum Anatomy 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 239000012149 elution buffer Substances 0.000 description 1
- 231100001135 endothelial toxicity Toxicity 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 238000013401 experimental design Methods 0.000 description 1
- 231100000573 exposure to toxins Toxicity 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 230000004761 fibrosis Effects 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 238000002073 fluorescence micrograph Methods 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 210000000232 gallbladder Anatomy 0.000 description 1
- 229960002518 gentamicin Drugs 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 108010087005 glusulase Proteins 0.000 description 1
- 230000002414 glycolytic effect Effects 0.000 description 1
- 239000005090 green fluorescent protein Substances 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 201000009277 hairy cell leukemia Diseases 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 239000000185 hemagglutinin Substances 0.000 description 1
- 230000009033 hematopoietic malignancy Effects 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 230000002440 hepatic effect Effects 0.000 description 1
- 230000002962 histologic effect Effects 0.000 description 1
- 239000000710 homodimer Substances 0.000 description 1
- 102000055277 human IL2 Human genes 0.000 description 1
- 210000004276 hyalin Anatomy 0.000 description 1
- 210000004408 hybridoma Anatomy 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 230000005965 immune activity Effects 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 230000003832 immune regulation Effects 0.000 description 1
- 208000026278 immune system disease Diseases 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 229940127121 immunoconjugate Drugs 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 229940097275 indigo Drugs 0.000 description 1
- COHYTHOBJLSHDF-UHFFFAOYSA-N indigo powder Natural products N1C2=CC=CC=C2C(=O)C1=C1C(=O)C2=CC=CC=C2N1 COHYTHOBJLSHDF-UHFFFAOYSA-N 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- ZPNFWUPYTFPOJU-LPYSRVMUSA-N iniprol Chemical compound C([C@H]1C(=O)NCC(=O)NCC(=O)N[C@H]2CSSC[C@H]3C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@H](C(N[C@H](C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC=4C=CC=CC=4)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CC=4C=CC=CC=4)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC2=O)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC=2C=CC=CC=2)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]2N(CCC2)C(=O)[C@@H](N)CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N2[C@@H](CCC2)C(=O)N2[C@@H](CCC2)C(=O)N[C@@H](CC=2C=CC(O)=CC=2)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N2[C@@H](CCC2)C(=O)N3)C(=O)NCC(=O)NCC(=O)N[C@@H](C)C(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@H](C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@H](C(=O)N1)C(C)C)[C@@H](C)O)[C@@H](C)CC)=O)[C@@H](C)CC)C1=CC=C(O)C=C1 ZPNFWUPYTFPOJU-LPYSRVMUSA-N 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 230000000968 intestinal effect Effects 0.000 description 1
- 210000005061 intracellular organelle Anatomy 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 210000001630 jejunum Anatomy 0.000 description 1
- 201000002215 juvenile rheumatoid arthritis Diseases 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 210000001865 kupffer cell Anatomy 0.000 description 1
- 210000002664 langerhans' cell Anatomy 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 231100000225 lethality Toxicity 0.000 description 1
- GDBQQVLCIARPGH-ULQDDVLXSA-N leupeptin Chemical compound CC(C)C[C@H](NC(C)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C=O)CCCN=C(N)N GDBQQVLCIARPGH-ULQDDVLXSA-N 0.000 description 1
- 108010052968 leupeptin Proteins 0.000 description 1
- 238000012417 linear regression Methods 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 230000002934 lysing effect Effects 0.000 description 1
- 238000002595 magnetic resonance imaging Methods 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 210000001161 mammalian embryo Anatomy 0.000 description 1
- 201000008350 membranous glomerulonephritis Diseases 0.000 description 1
- 238000001466 metabolic labeling Methods 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 230000002969 morbid Effects 0.000 description 1
- 230000036473 myasthenia Effects 0.000 description 1
- 206010028417 myasthenia gravis Diseases 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 210000005170 neoplastic cell Anatomy 0.000 description 1
- 238000006386 neutralization reaction Methods 0.000 description 1
- 231100000065 noncytotoxic Toxicity 0.000 description 1
- 230000002020 noncytotoxic effect Effects 0.000 description 1
- 210000000287 oocyte Anatomy 0.000 description 1
- 201000005737 orchitis Diseases 0.000 description 1
- 210000003463 organelle Anatomy 0.000 description 1
- 230000002611 ovarian Effects 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 208000003154 papilloma Diseases 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229950000964 pepstatin Drugs 0.000 description 1
- 108010091212 pepstatin Proteins 0.000 description 1
- FAXGPCHRFPCXOO-LXTPJMTPSA-N pepstatin A Chemical compound OC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C(C)C)NC(=O)[C@H](C(C)C)NC(=O)CC(C)C FAXGPCHRFPCXOO-LXTPJMTPSA-N 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 231100000614 poison Toxicity 0.000 description 1
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 238000003752 polymerase chain reaction Methods 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 238000011886 postmortem examination Methods 0.000 description 1
- 229910000160 potassium phosphate Inorganic materials 0.000 description 1
- 235000011009 potassium phosphates Nutrition 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 108010089682 preproricin Proteins 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 238000000164 protein isolation Methods 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- 235000021251 pulses Nutrition 0.000 description 1
- 238000011127 radiochemotherapy Methods 0.000 description 1
- 238000003127 radioimmunoassay Methods 0.000 description 1
- 239000009342 ragweed pollen Substances 0.000 description 1
- 230000010837 receptor-mediated endocytosis Effects 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000032537 response to toxin Effects 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 108020004418 ribosomal RNA Proteins 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 239000012723 sample buffer Substances 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 208000013220 shortness of breath Diseases 0.000 description 1
- 229910052710 silicon Inorganic materials 0.000 description 1
- 239000010703 silicon Substances 0.000 description 1
- 210000002027 skeletal muscle Anatomy 0.000 description 1
- 210000000278 spinal cord Anatomy 0.000 description 1
- 238000013125 spirometry Methods 0.000 description 1
- 238000012453 sprague-dawley rat model Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 230000003335 steric effect Effects 0.000 description 1
- 239000008174 sterile solution Substances 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- MNRILEROXIRVNJ-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=NC=N[C]21 MNRILEROXIRVNJ-UHFFFAOYSA-N 0.000 description 1
- 229960003087 tioguanine Drugs 0.000 description 1
- 230000024664 tolerance induction Effects 0.000 description 1
- 239000003440 toxic substance Substances 0.000 description 1
- 210000003437 trachea Anatomy 0.000 description 1
- 230000005030 transcription termination Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- KCOCBSPGEFYKMS-UHFFFAOYSA-K trisodium N,N-diethylethanamine phosphate Chemical compound P(=O)([O-])([O-])[O-].[Na+].C(C)N(CC)CC.[Na+].[Na+] KCOCBSPGEFYKMS-UHFFFAOYSA-K 0.000 description 1
- 125000001493 tyrosinyl group Chemical group [H]OC1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 241000712461 unidentified influenza virus Species 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- 210000004291 uterus Anatomy 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 230000003966 vascular damage Effects 0.000 description 1
- 108010043904 volkensin Proteins 0.000 description 1
- KQNNSYZQMSOOQH-GLDAUDTLSA-N volkensin Chemical compound C=1([C@@H]2C[C@@H]3O[C@@H](O)C[C@@H]4[C@]5(C)[C@H]6[C@H]([C@H]([C@@]4(C)C3=C2C)O)OC[C@]6(C)[C@H](OC(C)=O)C[C@@H]5OC(=O)C(/C)=C/C)C=COC=1 KQNNSYZQMSOOQH-GLDAUDTLSA-N 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/55—IL-2
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/415—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
-
- 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
- C07K14/475—Growth factors; Growth regulators
- C07K14/485—Epidermal growth factor [EGF], i.e. urogastrone
-
- 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
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/53—Colony-stimulating factor [CSF]
- C07K14/535—Granulocyte CSF; Granulocyte-macrophage CSF
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2809—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
Definitions
- the present invention relates to a ricin fusion protein immunotoxin and methods for treating a cancer or autoimmune disease with this immunotoxin.
- the present invention provides a ricin fusion protein immunotoxin comprising a ricin toxin B chain from which three lectin binding sites have been removed, conjugated to a ligand specific for a particular cell surface receptor and associated with a ricin toxin A chain.
- methods for administering the ricin fusion protein immunotoxin to treat a cancer or autoimmune disease in a patient diagnosed with a cancer or autoimmune disease are also provided.
- Ricin toxin a glycoprotein produced in the seeds ⁇ Ricinus communis plants consists of a galactose-binding B chain (RTB) disulfide linked to an rRNA N-glycosidase A chain (RTA).
- RTB galactose-binding B chain
- RTA rRNA N-glycosidase A chain
- the 65 kilodalton heterodimeric glycoprotein binds to cell surface galactose-terminated oligosaccharides via lectin binding sites in RTB (1) and undergoes receptor-mediated endocytosis (2). After trafficking to the Golgi (3), the toxin is transported to a distal compartment (4) from which the intersubunit disulfide bond is reduced (5).
- RTA then translocates to the cytosol and catalytically inactivates protein synthesis by hydrolysis of a specific adenine base from the 26S ribosomal RNA (6).
- Galactose binding is important for cell binding and may be needed for internalization and intracellular trafficking of ricin (34).
- Ricin toxin is one of the most toxic substances known to man. A single molecule is capable of causing cell death (7), and the LD 50 of ricin in 20 gram C57 B16 mice is reported to be 60 nanograms or 6 x 10 11 molecules (8). Histopathological examination of mice given toxic doses of ricin failed to show any definite abnormalities (8). Thus, the critical target organ for ricin is unknown.
- the x-ray crystallographic structure of ricin revealed two domains each with three subdomains each with similar folding and primary amino acid sequence (13). While all six subdomains had ⁇ -carbon chains forming a loop, twist and hook, only four of the subdomains (l ⁇ , l ⁇ , 2 ⁇ , and 2 ⁇ ) contained tripeptide kinks. Co-crystallization of 5 mM ⁇ -lactose with ricin showed sugar binding in the tripeptide kinks of subdomains l ⁇ and 2 ⁇ .
- Fusion toxins have been developed in an effort to treat these diseases. Fusion toxins are hybrid proteins composed of peptide ligands reactive with malignant cells (antibody fragments or cytokines) fused to polypeptide toxins [diphtheria toxin (DT), Pseudomonas exotoxin (PE) or ricin].
- DT diphtheria toxin
- PE Pseudomonas exotoxin
- ricin ricin
- the toxin- ligand-receptor complex internalizes into intracellular compartments from which the catalytic domain of the toxin translocates to the cytosol and inactivates protein synthesis.
- L2R interleukin-2 receptors
- IL2R bearing cells (31).
- Ricin' s extreme potency has led to its use in immunotoxins consisting of monoclonal antibodies chemically coupled to a modified ricin moiety.
- the large 200 kilodalton Mr immunoco ⁇ jugates showed significant vascular endothelial toxicity (32).
- Three groups of investigators have chemically or genetically modified lectin sites on ricin and used covalently attached ligands to study cell intoxication (33-35). In each case, reductions in lectin function led to profound decreases in cytotoxic potency.
- novel therapeutic modalities with minimal toxicities and no cross-resistance with current cytotoxic treatments are still needed.
- the present invention overcomes previous shortcomings by providing a ricin fusion protein immunotoxin comprising a ricin toxin B chain fusion protein having a modification in a lectin binding site in each of the l , l ⁇ and 2 ⁇ subunits and a ligand specific for a cell surface receptor in association with a ricin toxin A chain that selectively targets and intoxicates very specific cell populations for the treatment of cancer and autoimmune disease.
- the present invention provides a plant holotoxin comprising 1 ⁇ , 1 ⁇ and 2 ⁇ subdomains and having a modification in a lectin binding site in each of the
- a ricin toxin B chain having a modification in a lectin binding site in each of the l , l ⁇ and 2 ⁇ subdomains is provided.
- the present invention provides a plant holotoxin fusion protein comprising a moiety consisting of a plant holotoxin comprising l ⁇ , l ⁇ and 2 ⁇ subdomains and having a modification in a lectin binding site in each of the l ⁇ , l ⁇ and 2 ⁇ subdomains and a moiety consisting of a ligand specific for a cell surface receptor.
- a ricin toxin B chain fusion protein comprising a moiety consisting of a ricin toxin B chain having a modification in a lectin binding site in each of the l ⁇ , l ⁇ and 2 ⁇ subdomains and a moiety consisting of a ligand specific for a cell surface receptor is provided.
- a method of constructing a ricin fusion protein immunotoxin comprising expressing the nucleic acid in a vector in a eukaryotic cell expression system to produce a fusion protein; isolating and purifying the fusion protein; and contacting the fusion protein of with a ricin toxin A chain under conditions which permit the association of the fusion protein with the ricin toxin A chain.
- the present invention provides a method of treating a cancer or an autoimmune disease in a patient diagnosed with a cancer or an autoimmune disease comprising constructing a ricin fusion protein immunotoxin, wherein the ligand is specific for a particular cell surface receptor present only on the surfaces of the cancer cells or on the surfaces of the cells causing the patient's autoimmune disease; and administering the ricin fusion protein immunotoxin in a pharmaceutically acceptable carrier to the patient, whereby the ricin fusion protein immunotoxin treats the patient's cancer or autoimmune disease.
- a ricin protein immunotoxin comprising a ricin toxin A chain associated with a ricin toxin B chain fusion protein comprising a ricin toxin B chain having a W to S substitution at amino acid position 37 in the l ⁇ subdomain, a Y to H substitution at amino acid position 248 in the 1 ⁇ subdomain and a Y to H substitution at position 78 in the 2 ⁇ subdomain and a ligand specific for a cell surface receptor.
- LL2R is a heterotrimeric glycoprotein complex on the cell membrane with a 55 kD ⁇ subunit, a 75 kD ⁇ subunit and a 64 kD ⁇ subunit (16)
- the only normal human tissues expressing IL2R ⁇ and IL2R ⁇ are activated T cells, B cells, LGL cells and monocytes and some liver Kupffer cells, lung macrophages and skin Langerhans' cells
- an immunotoxin targeted to this receptor is expected to be reasonably selective
- a variety of hematologic neoplasms may show high affinity LL2R expression including hairy cell leukemia, adult T cell leukemia and a fraction of cutaneous T cell lymphomas and B-cell chronic lymphocytic leukemias (17)
- DT and PE have been fused to either LL2 or antibody Fv anti-IL2R peptides (18-25) All reagents showed potent selective cytoplasm
- Ricin-based fusion proteins are attractive candidates for development for several reasons
- the toxin inactivates cell protein synthesis by a mechanism independent of that used by DT or PE
- the RNA N-glycosidase activity of ricin cripples 1500 ribosomes/minute and a single molecule of ricin in the cytosol can cause cell death (6,27)
- ricin fusion toxins may be used in combination with bacterial fusion toxins or when bacterial fusion toxin resistance is encountered
- there is no immunologic cross- reactivity between ricin and the bacterial toxins Patients who have been immunized with DT or had previous exposure to PE do not show amnestic immune responses to ricin (28)
- the present invention provides a plant holotoxin comprising 1 ⁇ , 1 ⁇ and 2 ⁇ subdomains and having a modification in a lectin binding site in each of the
- the plant holotoxin can be, but is not limited to, ricin, mistletoe toxin, abrin, volkensin as well as any other compound now known or identified in the future to be a plant holotoxin
- the preferred plant holotoxin will be characterized as a 65 kD glycoprotein with a galactose lectin B chain and RNA N-glycosidase A chain
- the plant holotoxin with the modification described herein can have various functional attributes, such as, for example, the ability to be produced in high yields as an expressed protein in a eukaryotic gene expression system, specific reactivity with antibodies against the plant holotoxin, proper folding to retain the stability and functional characteristics of the wild type holotoxin, proper association with additional moieties which are normally associated with the wild type holotoxin, a 50% lethal dose (LD 50 ) value of greater than ten micrograms in mice, at least a one thousand fold reduction in sugar binding as
- LD 50 50% lethal dose
- the modification in a lectin binding site in each of the 1 ⁇ , 1 ⁇ and 2 ⁇ subdomains of the plant holotoxin can be an amino acid substitution, such as, for example, a substitution of an amino acid having an aromatic ring residue with an amino acid lacking an aromatic ring residue
- Other modifications in the lectin binding sites can include, but are not limited to, for example, substitution of an amino acid with a polar hydrogen binding residue with an amino acid with a nonpolar residue, as well as other deletions, additions or amino acid substitutions or any other modifications now known or later discovered that result in either complete or significant removal of the sugar binding activity of the lectin binding site.
- the plant holotoxin with modifications will maintain a functional conformation and be able to associate normally with the intoxication-imparting moiety. Whether a given modification results in the complete or significant removal of the sugar binding activity of a lectin binding site can be determined according to the protocols provided in the Examples herein.
- a ricin toxin B chain having a modification in a lectin binding site in each of the l , l ⁇ and 2 ⁇ subdomains.
- the ricin toxin B chain with such modification can have various functional attributes, such as, for example, the ability to be produced in high yields as an expressed protein in a eukaryotic gene expression system; specific reactivity with antibodies against the ricin toxin B chain; proper folding to retain the stability and functional characteristics of the wild type ricin toxin B chain; proper association with ricin toxin A chain; a 50% lethal dose (LD 50 ) value of greater than ten micrograms in mice; at least a one thousand fold reduction in sugar binding as compared with wild type ricin toxin B chain, at least a one hundred fold reduction in toxicity in mice as compared with wild type ricin toxin B chain; and the ability to selectively intoxicate a target cell with a two hundred fold reduction in the ability to intoxicate a non-
- the modification in a lectin binding site in each of the l , l ⁇ and 2 ⁇ subdomains in the ricin toxin B chain of this invention can be an amino acid substitution, such as, for example, wherein the amino acid substitution consists of substitution of an amino acid having an aromatic ring residue with an amino acid lacking an aromatic ring residue.
- Other modifications in the lectin binding sites can include, but are not limited to, for example, substitution of an amino acid with a polar hydrogen binding residue with an amino acid with a nonpolar residue, as well as other deletions, additions or amino acid substitutions or any other modifications now known or later discovered that result in either complete or significant removal of the sugar binding activity of the lectin binding site. Whether a given modification results in the complete or significant removal of the sugar binding activity of a lectin binding site can be determined according to the protocols provided in the Examples herein.
- the amino acid substitutions can consist of a W to S substitution at amino acid position 37 in the l ⁇ subdomain, a Y to H substitution at amino acid position 248 in the 1 ⁇ subdomain and a Y to H substitution at position 78 in the 2 ⁇ subdomain.
- Other modifications in the lectin binding sites can include, but are not limited to, for example, substitution of an amino acid with a polar hydrogen binding residue with an amino acid with a nonpolar residue, as well as other deletions, additions or amino acid substitutions or any other modifications now known or later discovered that result in either complete or significant removal of the sugar binding activity of the lectin binding site. Whether a given modification results in the complete or significant removal of the sugar binding activity of a lectin binding site can be determined according to the protocols provided in the Examples herein.
- a plant holotoxin comprising 1 ⁇ , 1 ⁇ and 2 ⁇ subdomains and having a modification in a lectin binding site in one or more or the subdomains, including at least a modification in the 1 ⁇ subdomain is also contemplated in the present invention.
- the modification in the lectin binding site in one or more of the subdomains can be an amino acid substitution, such as for example, a substitution of an amino acid having an aromatic ring residue with an amino acid lacking an aromatic ring residue (e.g., a Y to H substitution at amino acid position 248 in the 1 ⁇ subdomain).
- a ricin toxin B chain fusion protein having a modification in a lectin binding site in one or more subdomain, including at least a modification in the 1 ⁇ subdomain.
- the modification in one or more of the subdomains can be an amino acid substitution, such as, for example, a substitution of an amino acid having an aromatic ring residue with an amino acid lacking an aromatic ring residue.
- the amino acid substitution can be, but is not limited to, a Y to H substitution at position 248 in the 1 ⁇ subdomain.
- a plant holotoxin fusion protein comprising a plant holotoxin comprising l ⁇ , l ⁇ and 2 ⁇ subdomains and having a modification in a lectin binding site in each of the l ⁇ , l ⁇ and 2 ⁇ subdomains and a ligand specific for a cell surface receptor.
- the ligand of the plant holotoxin fusion protein can be, but is not limited to, interleukin-2, granulocyte/macrophage colony stimulating factor, an antibody or antibody fragment to CD3, an antibody or antibody fragment to GD2, epidermal growth factor, IGF2, GRF, substance P, MSH, as well as any other molecular entity now known or identified in the future to be a ligand specific for a cell surface receptor as determined by assaying a potential ligand for selective binding avidity for a particular cell surface receptor by protocols standard in the art for measuring binding avidities.
- the present invention additionally provides a ricin toxin B chain fusion protein comprising a ricin toxin B chain having a modification in a lectin binding site in each of the l ⁇ , l ⁇ and 2 ⁇ subdomains as described above and a ligand specific for a cell surface receptor.
- the ligand of the ricin toxin B chain fusion protein can be, but is not limited to, interleukin-2, granulocyte/macrophage colony stimulating factor, an antibody to CD3, an antibody to GD2, epidermal growth factor, an antibody or antibody fragment to CD3, an antibody or antibody fragment to GD2, epidermal growth factor, IGF2, GRF, substance P, MSH, as well as any other molecular entity now known or identified in the future to be a ligand specific for a cell surface receptor as determined by assaying a potential ligand for selective binding avidity for a particular cell surface receptor by protocols standard in the art for measuring binding avidities.
- the present invention also provides a ricin toxin B chain fusion protein comprising a ricin toxin B chain having a W to S substitution at amino acid position 37 in the l ⁇ subdomain, a Y to H substitution at amino acid position 248 in the 1 ⁇ subdomain and a Y to H substitution at position 78 in the 2 ⁇ subdomain and a ligand specific for a cell surface receptor.
- a ricin fusion protein immunotoxin comprising this ricin toxin B chain fusion protein associated with a ricin toxin A chain.
- a plant holotoxin fusion protein immunotoxin is also provided in the present invention, comprising a plant holotoxin fusion protein (which imparts a binding function to the immunotoxin) as described above, associated with a moiety imparting an intoxicating function to the immunotoxin.
- a ricin fusion protein immunotoxin is provided in the present invention, comprising a ricin toxin B chain fusion protein consisting of a ricin toxin B chain having a modification in a lectin binding site in each of the l ⁇ , l ⁇ and 2 ⁇ subdomains and a ligand specific for a cell surface receptor, associated with a ricin toxin A chain.
- Nucleic acids encoding the plant holotoxins and fusion proteins of this invention are also contemplated, as well as vectors comprising the nucleic acids and hosts comprising the vectors.
- the present invention also provides nucleic acids complementary to or capable of hybridizing with the nucleic acids encoding the plant holotoxins and fusion proteins.
- the nucleic acid of the plant holotoxin or fusion protein can encode the intact plant holotoxin with the modifications described herein or an active fragment thereof.
- An active fragment of a plant holotoxin is a fragment which is capable of maintaining a functional conformation and associating with the moiety which imparts an intoxicating function to the plant holotoxin.
- the nucleic acid of the fusion protein can also encode the intact ligand or an active fragment thereof.
- An active fragment of a ligand of this invention is a fragment which is capable of maintaining a functional conformation and specifically binding to its corresponding cell surface receptor.
- Protocols for construction of a vector containing a nucleic acid encoding a plant holotoxin fusion protein such as the ricin toxin B chain fusion protein are well known in the art and are described in the Examples provided herein.
- the vector can be expressed in any in vitro eukaryotic cell expression system, such as, for example, the Spodoptera frugiperda insect cell line which expresses proteins in a baculovirus vector, as described in the Examples herein.
- Isolation and purification of the expressed fusion protein can be carried out by protocols well known to those of skill in the art, e.g., as described in the Examples herein.
- a method of constructing a ricin fusion protein immunotoxin comprising expressing a nucleic acid encoding the ricin toxin B fusion protein, in a vector in a eukaryotic cell expression system, to produce a ricin toxin B chain fusion protein; isolating and purifying the ricin toxin B chain fusion protein; and contacting the ricin toxin B chain fusion protein with a ricin toxin A chain under conditions which permit the association of the ricin toxin B chain fusion protein with the ricin toxin A chain.
- the present invention further contemplates a method of constructing a plant holotoxin fusion protein immunotoxin comprising expressing a nucleic acid encoding the plant holotoxin fusion protein described above, in a vector in a eukaryotic cell expression system to produce a plant holotoxin fusion protein; isolating and purifying the plant holotoxin fusion protein; and contacting the plant holotoxin fusion protein with a moiety which imparts an intoxicating function, under conditions which permit the association of the plant holotoxin fusion protein with the intoxicating moiety to yield a plant holotoxin fusion protein immunotoxin.
- vectors and expression systems described in the Examples herein a variety of vectors and eukaryotic expression systems such as yeast, filamentous fungi, insect cell lines, bird, fish, transgenic plants and mammalian cells, among others, as are known to those of ordinary skill in the art, can also be used in the present invention.
- the vectors of the invention can be in a host (e.g., cell line or transgenic animal) that can express the nucleic acid contemplated by the present invention.
- a host e.g., cell line or transgenic animal
- the vector e.g., a plasmid, which is used to transform the host cell, preferably contains DNA sequences to initiate transcription and sequences to control the translation of the protein. These sequences are referred to as expression control sequences.
- Suitable vectors for expression systems usually have expression control sequences, such as promoters, including 3-phosphoglycerate kinase or other glycolytic enzymes, an origin of replication, termination sequences and the like, as desired.
- Preferred expression control sequences are promoters derived from immunoglobulin genes, SV40 virus, adenovirus, bovine papilloma virus, etc, as are well known in the art.
- gene sequences to control replication in the host cell may be incorporated into the vector such as those found in bovine papilloma virus-type vectors (64).
- suitable vectors are described in the literature (see, for example, 65,66).
- Appropriate vectors for expressing proteins in insect cells are usually derived from baculovirus.
- suitable insect cell lines include, but are not limited to, mosquito larvae, silkworm, armyworm, moth and Drosophila cell lines such as a Schneider cell line (67), as well as any other insect cell line now known or identified in the future to be a suitable host cell line for baculovirus or other insect cell expression vectors.
- polyadenylation or transcription terminator sequences from known mammalian genes can be incorporated into the vector.
- An example of a terminator sequence is the polyadenylation sequence from the bovine growth hormone gene. Sequences for accurate splicing of the transcript may also be included.
- An example of a splicing sequence is the VP1 intron from SV40 (68).
- the nucleic acid sequences can be expressed in hosts after the sequences have been operably linked to, i.e., positioned, to ensure the functioning of an expression control sequence.
- These expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA.
- expression vectors can contain selection markers, e.g., tetracycline resistance, hygromycin resistance, gentamicin resistance or methotrexate resistance, to permit detection and/or selection of those cells transformed with the desired nucleic acid sequences (see, e.g., U.S. Patent 4,704,362).
- the presence of the vector RNA in transformed cells can be confirmed by Northern blot analysis and production of a cDNA or opposite strand RNA corresponding to the antigen coding sequence can be confirmed by Southern and Northern blot analysis, respectively.
- Polynucleotides encoding a variant polypeptide may include sequences that facilitate transcription (expression sequences) and translation of the coding sequences such that the encoded polypeptide product is produced. Construction of such polynucleotides is well known in the art. For example, such polynucleotides can include a promoter, a transcription termination site (polyadenylation site in eukaryotic expression hosts), a ribosome binding site and optionally, an enhancer for use in eukaryotic expression hosts as well as any sequences necessary for replication of a vector.
- the host cells are rendered competent for transformation by various means known in the art. There are several well-known methods of introducing
- DNA into eukaryotic cells include, but are not limited to, calcium phosphate precipitation, fusion of the recipient cells with bacterial protoplasts containing the DNA, treatment of the recipient cells with liposomes containing the DNA DEAE dextran, electroporation, micro-injection of the DNA directly into the cells, as well as any other technique now known or developed in the future for introducing nucleic acid into cells.
- the transformed cells are cultured by means well known to one of ordinary skill in the art (69).
- the expressed polypeptides are isolated from cells grown as suspensions or monolayers. The latter are recovered by well- known mechanical, chemical, or enzymatic means and purified according to standard methods well known in the art.
- yeast cells are first converted into protoplasts using zymolase, lytiacase, or glusulase, followed by addition of DNA and polyethylene glycol (PEG).
- PEG polyethylene glycol
- the PEG- treated protoplasts are then regenerated in a 3% agar medium under selective conditions. Details of this procedure are described by Beggs, J.D. (71) and Hinnen et al. (72).
- the second procedure does not involve removal of the cell wall. Instead, the cells are treated with lithium chloride or acetate and PEG and put on selective plates (73).
- Plant holotoxins and fusion proteins once expressed, can be isolated from yeast by lysing the cells and applying standard protein isolation and purification techniques to the lysates.
- the monitoring of the purification process can be accomplished by using Western blot techniques or radioimmunoassay or other standard immunoassay techniques.
- sequences encoding plant holotoxins and fusion proteins of the present invention can also be ligated to various expression vectors for use in transforming cell cultures of, for example, mammalian, insect, plant, bird or fish origin.
- Illustrative of cell cultures useful for the production of polypeptides are mammalian cells. Mammalian cells permit the expression of proteins in an environment that favors important post-translational modifications such as folding and cysteine pairing, addition of complex carbohydrate structures and secretion of active protein.
- Mammalian cell systems can be in the form of monolayers of cells, although mammalian cell suspensions may also be used.
- a number of suitable host cell lines capable of expressing intact proteins have been developed in the art, and include the HEK293, BHK21 and CHO cell lines, as well as various human cells such as COS cell lines, HeLa cells, myeloma cell lines, Jurkat cells, etc., as are known in the art.
- Other animal cells useful for the production of proteins are available, for example, from the American Type Culture Collection
- Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter (e.g., the CMV promoter, a HSC tk promoter or pgk [phosphoglycerate kinase] promoter), an enhancer (74) and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.g., an SV40 large T antigen poly A addition site) and transcriptional terminator sequences.
- a promoter e.g., the CMV promoter, a HSC tk promoter or pgk [phosphoglycerate kinase] promoter
- an enhancer e.g., the CMV promoter, a HSC tk promoter or pgk [phosphoglycerate kinase] promoter
- necessary processing information sites such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.g., an
- Efficient post translational glycosylation and expression of recombinant proteins can also be achieved in insect cell expression systems employing baculovirus vectors, as described in the Examples herein.
- the plant holotoxin of this invention can also be expressed in transgenic plant expression systems known in the art, such as, for example, soy bean cells or Nicotiana tabacum cells (83).
- the nucleic acids of the present invention can be used to generate transgenic nonhuman animals in which the nucleic acid encoding a plant holotoxin or fusion protein of the present invention is added to the germ line of the animal.
- a cell of the invention containing an nucleic acid of this invention is contemplated to include a cell in a transgenic animal.
- the plant holotoxin or fusion protein can be isolated and purified from materials secreted by the animal, such as for example, milk secreted from nonhuman mammals.
- Transgenic animals are generated by standard means known to those skilled in the art (see, for example, 84)
- Ricin toxin A chain can be obtained commercially or by expression of a nucleic acid encoding ricin toxin A chain in prokaryotic or eukaryotic in vitro expression systems according to standard protocols known in the art and associated with the ricin toxin B chain fusion protein to produce the immunotoxin (82).
- the nucleic acid encoding the ricin toxin A chain can also be included in a vector which also comprises the nucleic acid encoding the ricin toxin B chain fusion protein of this invention. This vector can be introduced into a eukaryotic expression system, such as a plant cell expression system
- the terms “selective” or “selectively” and “specific” or “specifically” all have the same meaning and are thus used interchangeably to mean either that the ligand binds to only one type of cell surface receptor which has been identified as binding that particular ligand and does not randomly bind to other cell surface molecules, that the antibody binds its corresponding antigen which has been identified and does not randomly bind with other antigens, or that the immunotoxin binds its intended target cell which has been identified and does not randomly bind to other nontarget cells.
- Also contemplated for the present invention is a method for treating or preventing a cancer or an autoimmune disease in a patient comprising constructing the plant holotoxin fusion protein immunotoxin of the present invention as described above, wherein the ligand is specific for a particular cell surface receptor present on the surfaces of the cells to be targeted for intoxication and killing for the purpose of treating or preventing a cancer or autoimmune disease; and administering the plant holotoxin fusion protein immunotoxin in a pharmaceutically acceptable carrier to the patient, whereby the plant holotoxin fusion protein immunotoxin treats or prevents a cancer or autoimmune disease in the patient.
- the present invention provides a method of treating or preventing a cancer or an autoimmune disease in a patient comprising constructing the ricin fusion protein immunotoxin of the present invention as described above, wherein the ligand is specific for a particular cell surface receptor present on the surfaces of the cells to be targeted for intoxication and killing; and administering the ricin fusion protein immunotoxin in a pharmaceutically acceptable carrier to the patient, whereby the ricin fusion protein immunotoxin treats or prevents a cancer or autoimmune disease in the patient.
- the treatment or prevention of the cancer or autoimmune disease is by the specific intoxication and killing of the cells associated, or potentially associated, with a cancer or autoimmune disease, resulting in prevention of or remission of the cancer, or prevention, elimination or reduction in severity of the symptoms of autoimmune disease.
- That a given plant holotoxin fusion protein immunotoxin is effective in treating or preventing a cancer or autoimmune disease in a patient can be determined by evaluating the particular aspects of the medical history, the signs, symptoms and objective laboratory tests that have a documented utility in evaluating disease activity. These signs, symptoms and objective laboratory tests will vary depending on the particular cancer or autoimmune disease being treated or prevented as will be well known to any clinician in this field.
- Such methods can include, but are not limited to, x-rays, biopsies of biological samples, palpation of masses and measurements of blood and body fluid components.
- autoimmune disease e.g., multiple sclerosis
- clinical parameters that can be monitored can include the severity and number of attacks, or for continuously progressive disease, the worsening of symptoms and signs, the cumulative development of disability, the number or extent of brain lesions as determined by magnetic resonance imaging and the need for continued use of immunosuppressive medications (78,79).
- the cancer to be treated or prevented by administration of the plant holotoxin fusion protein immunotoxin can be, but is not limited to, a human leukemia or lymphoma having cancer cells expressing interleukin-2 receptors on the surfaces, wherein the ligand is interleukin-2, acute myelogenous leukemia, wherein the ligand is granulocyte/macrophage-colony stimulating factor and melanoma/neuroblastoma, wherein the ligand is an antibody to GD2, as well as brain neoplasms, epithelial malignancies, sarcomas or any other cancer now known or identified in the future which can be treated or prevented by administration of the plant holotoxin fusion protein immunotoxin of the present invention.
- a human leukemia or lymphoma having cancer cells expressing interleukin-2 receptors on the surfaces, wherein the ligand is interleukin-2, acute myelogenous leukemia, wherein the ligand is
- Such a cancer would express on the surface of the tumor cells or potential tumor cells an antigen to which the ligand of the present immunotoxin can selectively bind, for selective intoxication of the tumor cells or potential tumor cells by the ricin protein immunotoxin
- an antigen to which the ligand of the present immunotoxin can selectively bind for selective intoxication of the tumor cells or potential tumor cells by the ricin protein immunotoxin
- tumor antigens or potential tumor antigens such as, but not limited to, growth factor receptors, adhesion molecules, oncogene products, differentiation antigens and oncofetal antigens on the surface of a subject's tumor cells or potential tumor cells
- autoimmune disease describes a disease state or syndrome whereby a subject's body produces a dysfunctional immune response against the subject's own body components, with adverse effect
- the autoimmune disease to be treated or prevented by administration of the ricin protein immunotoxin of the present invention can be, but is not limited to, graft-versus-host disease, wherein the ligand can be an antibody to CD3
- Other examples of autoimmune diseases that can be treated or prevented include ulcerative colitis, Crohn's disease, multiple sclerosis, rheumatoid arthritis, diabetes mellitus, pernicious anemia, autoimmune gastritis, psoriasis, Bechet's disease, idiopathic thrombocytopenic purpura, Wegener's granulomatosis, autoimmune thyroiditis, autoimmune oophoritis, bullous pemphigoid, pemphigus, polyendocrinopathies, Still's disease, Lambert-Eaton myasthenia syndrome, myas
- allergic disease describes a disease state or syndrome whereby the body produces a dysfunctional immune response composed of IgE antibodies to environmental antigens and which evoke allergic symptoms.
- allergic diseases include, but are not limited to, asthma, ragweed pollen hayfever, allergy to food substances and allergic reactions.
- the treatment or prevention of the allergic disease is by the specific intoxication and killing of the cells associated with ,or potentially associated with, the allergic disease, resulting in the prevention of symptoms or the elimination or reduction in severity of the symptoms of the allergic disease. That a given plant holotoxin fusion protein immunotoxin is effective in treating or preventing an allergic disease in a patient can be determined by evaluating the particular aspects of the medical history, the signs, symptoms and objective laboratory test that have a documented utility in evaluating disease activity.
- allergic diseases can include, but are not limited to, x-rays, biopsies of biological samples, palpation of masses and measurements of blood and other body fluid components.
- clinical parameters that can be monitored for an allergic disease e.g., asthma
- the measurement of airway resistance by the use of respiratory spirometry, the extent of disability and the dependence on immunosuppressive medications or bronchodilators can also be determined (80,81).
- Also contemplated for the present invention is a method of inducing immune tolerance in the patient, comprising constructing the plant holotoxin fusion protein immunotoxin of the present invention as described above, wherein the ligand is specific for a particular cell surface receptor present on the surfaces of certain immune cells to be targeted for intoxication and killing; and administering the plant holotoxin fusion protein immunotoxin in a pharmaceutically acceptable carrier to the patient, whereby the plant holotoxin fusion protein immunotoxin induces immune tolerance.
- the ligand can be IL2 or an antibody or fragment thereof (e.g., Fv region) to the CD3 antigen.
- the induction of immune tolerance is by the specific intoxication and killing of certain immune cells, resulting in the elimination or reduction in severity of a particular immune response. That a given plant holotoxin fusion protein immunotoxin is effective in inducing an immune tolerance in a patient can be determined by evaluating the particular aspects of the medical history, the signs, symptoms and objective laboratory tests that have a documented utility in evaluating immune activity. These signs, symptoms and objective laboratory tests will vary depending on the particular immune response being reduced or eliminated, as will be well known to any clinician in this field. Examples of such methods include, but are not limited to, x-rays, biopsies of biological samples, palpation of masses and measurements of blood and other body fluid components. In particular, cytokine assays, routine clinical chemistries, immune function assays, complete blood counts and the like, as would be known to the clinician, can be measured at various intervals during treatment.
- immune tolerance e.g., transplantation rejection
- This time interval may be large, with respect to the development of cancer, autoimmune or allergic diseases (years/decades) or short (weeks/months) with respect to the development of a need for induction of immune tolerance.
- the determination of who would be at risk for the development of a cancer, autoimmune disease, allergic disease or in need of induction of immune tolerance would be made based on current knowledge of the known risk factors for a particular disease or immune response familiar to a clinician in this field, such as a particularly strong family history of disease or need for a transplant.
- the plant holotoxins, fusion proteins and immunotoxins of the present invention are preferably provided in a pharmaceutically acceptable carrier and can be parenterally administered to the subject.
- Suitable carriers for parenteral administration of the immunotoxin in a sterile solution or suspension can include sterile saline that may contain additives, such as ethyl oleate or isopropyl myristate, and can be injected, for example, intravenously, as well as into subcutaneous or intramuscular tissues.
- the plant holotoxin fusion protein immunotoxin can be administered to the subject in amounts sufficient to treat or prevent a cancer, autoimmune disease or allergic disease, or to induce immune tolerance. Optimal dosages used will vary according to the individual, as well as the particular cancer or autoimmune or allergic disease being treated or the type of immune response being induced. Typically, for treatment of humans, plant holotoxin fusion protein immunotoxin, (e.g., ricin toxin fusion protein immunotoxin) would be administered intravenously in a dosage range between 1 ⁇ g and 10 mg/kg of body weight and most preferably in a dose of 0.5 mg/kg, either as a single bolus or as a continuous infusion ranging in time from a day to a month. Treatment can be continued for an indefinite period of time, as indicated by monitoring of the signs, symptoms and clinical parameters associated with a particular cancer, autoimmune disease, allergic disease or immune response induction.
- plant holotoxin fusion protein immunotoxin e.g., ricin
- the amount of plant holotoxin fusion protein immunotoxin administered will also vary among individuals on the basis of age, size, weight, condition, etc.
- dosages are best optimized by the practicing physician and methods for determining dosage are described, for example, in Remington 's Pharmaceutical Sciences (11). That a given dosage amount or regimen is effective in treating a cancer, autoimmune disease, or allergic disease or inducing immune tolerance, can be readily determined by using the parameters described above.
- the present invention is more particularly described in the following examples which are intended as illustrative only since numerous modifications and variations therein will be apparent to those skilled in the art.
- EXAMPLE I Construction of a ricin toxin containing a modification in a lectin binding site in each of the l ⁇ , l ⁇ and 2 ⁇ subdomains.
- DNAs (4 ⁇ g) were co-transfected with 0.5 ⁇ g of BaculoGold AcNPV DNA (PharMingen) into 2 x 10 6 Sf9 Spodoptera frugiperda insect cells as recommended by the supplier.
- media were centrifuged and supernatants tested in limiting dilution assays with Sf9 cells as previously described (40). Positive wells were identified and supernatants reassayed by limiting dilution until all wells up to 10 "8 dilution were positive. Two rounds of selection were required for each mutant.
- Recombinant viruses in the supernatants were then amplified by infecting Sf9 cells at a multiplicity of infection (moi) of 0.1 , followed by collection of day 7 supernatants.
- moi multiplicity of infection
- mutant RTBs Recombinant baculoviruses were used to infect Sf9 cells at an moi of 5 in EX-CELL400 media (JRH Scientific, Lexena, KS) with 25 mM ⁇ -lactose in spinner flasks. Media supernatants and cell pellets containing mutant RTBs were collected day
- Cell pellets were dissolved in 20 mM Tris HCL( pH 8), 50 mM NaCl, 1% NP40, 1 mM PMSF, 2 ug/ml aprotinin, 1.5 ug/ml pepstatin and 1.5 ug/ml leupeptin, frozen at -70°C, thawed, centrifuged at 22,000g for 15 minutes at 4°C , dialyzed into NTEAL at 4°C and treated identically to dialyzed concentrated cell supernatants.
- mutant RTBs Reassociation of mutant RTBs with plant RTA to form heterodimers.
- 0.25 ml of mutant RTBs (5 - 15 ⁇ g) was mixed with a 4 - fold molar excess of plant RTA (Inland Laboratories) in 0.1 M triethylamine-sodium phosphate pH 7 overnight at room temperature.
- the reaction mixture was then analyzed by a ricin ELISA utilizing P2 monoclonal anti-RTB coated wells, biotin conjugated ⁇ BR12 monoclonal anti-RTA and alkaline phosphatase-conjugated streptavidin detection reagents, as previously described (15).
- W37S/Y248H/W160S RTB The yields were not dissimilar from the yields for the double-site mutant W37S/Y248H (220 ug/liter from supernatant and 250 ug/liter from cell pellet) or wild-type RTB (400 ug/liter for supernatants) in this expression system and may reflect proper folding for the triple-site mutants.
- the conservative modification of surface residues may have contributed to protein stability.
- the double-site RTB mutant, W37S/Y248H, bound asialofetuin 4.8 ⁇ 2% (n ⁇ ) relative to recombinant or plant RTB.
- the triple-site RTB mutant, W37S/Y248H/W160S, bound asialofetuin similarly at 1.1 ⁇ 0.27% (n 7) relative to plant RTB.
- mutant RTB binding to glycoproteins was made by detecting mutant RTB bound to cell surfaces. Only W37S/Y248H and W37S/Y248H/W160S showed significant binding to KB cells at 4°C. Sugar binding of triple-site mutants. W37S/Y248H/W160S RTB retained binding to immobilized asialofetuin and KB cell surface glycoproteins. In both cases, the binding was competed with soluble saccharides. In contrast, W37S/Y248H/Y78H had minimal to negligible sugar binding.
- the IC 50 of ricin on HUT 102 human leukemia cells was 4 x 10 '12 M.
- the IC 50 for W37S/Y248H was 2 x 10 "10 M; the IC 50 for W37S/Y248H/W160S was 1 x 10 "10 M; and the IC 50 for W37S/Y248H/Y78H was 5 x 10 "9 M.
- Plant RTA alone had a 20-fold higher
- ricin lectin sites Widely separated ricin lectin sites.
- the 1 ⁇ and 2 ⁇ sites are separated by 36 Angstroms, l ⁇ and 2 ⁇ sites are 44 Angstroms apart.
- the l ⁇ and l ⁇ distance is 19 Angstroms.
- These inter-binding site distances are much larger than the inter-site spacing for the hepatic Gal/GalNac receptor and its triantennary N-glycoside ligand (10 -20 Angstroms) (41).
- the ricin geometry resembles the spacing of sites on surface binding lectins including mammalian mannose-binding protein, influenza virus hemagglutinin, pertussis toxin and cholera toxin (42).
- proteins are phylogenetically unrelated based on lack of primary or tertiary structure homology. Nevertheless, in all these proteins, the sugar combining sites are multiple widely spaced and project toward a single plane. Thus, they are ideally suited for binding to eukaryotic cell surfaces.
- the three binding sites on ricin may provide the optimal geometry for binding to the uneven galactosyl oligosaccharide-rich surface of mammalian cells similar to camera tripods or stools.
- the RTB l ⁇ and 2 ⁇ subdomains are unlikely to contribute additional sugar binding as they lack the tripeptide ⁇ -carbon kink, aromatic residues or charged residues for hydrogen bond formation. Further, no RGD-like domains exist in RTB—unlike discoidin I from the slime mold, Dictylostelium discoideum (46).
- EXAMPLE II In vivo toxicity of a ricin toxin containing a modification in a lectin binding site in each of the l ⁇ , l ⁇ and 2 ⁇ subdomains.
- Ricin and recombinant heterodimers Purified castor bean ricin (3.9 mg/ml in 0.15 M NaCl, 0.015 M potassium phosphate pH 7, 0.1% sodium azide ) was purchased from Sigma (St. Louis, MO). Purified deglycosylated RTA (5 mg/ml in PBS) was a gift of Dr. Jerry Fulton, Inland Laboratories, Dallas, Texas. Partially purified recombinant mutant RTBs were prepared and reassociated with plant RTA as previously described (15,40). The proteins tested and their characteristics are shown in Table 1. Ricin was stored at 4°C and all other tested proteins were stored at -20°C until used.
- mice Pathogen-free C57B/6 female mice (16-18 grams) were purchased from Jackson Laboratories (Bar Harbor, ME) or Harlan Sprague- Dawley (Indianapolis, IN), culled on each experiment to obtain 24 animals each
- mice 18 + 1 grams, and housed in groups of four in specific pathogen-free environment in Micro-Isolator cages (Lab Products, Maywood, NY).
- Experimental Design Mice were injected intraperitoneally with dilutions of toxins in phosphate buffered saline (PBS) plus 0 5% bovine serum albumin (BSA) so that each animal received 0 25 - 0 5 milliliters solution Animals were then observed twice daily for mortality Six different concentrations were tested on groups of four mice each for each protein Graphs of animal survival versus time were prepared for each protein Once the LD 50 was determined for each protein, single animals received the LD 50 and were collected at death or when severely morbid and autopsies were performed
- PBS phosphate buffered saline
- BSA bovine serum albumin
- mice Postmortem examinations were carried out on mice given LD 50 doses on moribund animals
- Samples from lungs, thymus, heart, esophagus, trachea, lymph node, liver, spleen, pancreas, kidneys, adrenals, gallbladder, stomach, duodenum, jejunum, colon, ovaries, uterus, brain, spinal cord, and skeletal muscle were taken for microscopic examination
- the tissues were fixed in 4% buffered formaldehyde and embedded in paraffin Sections were stained with hematoxylin and eosin
- the 1 ⁇ g dose killed two mice— one each on day 4 and 5
- the 500 ng dose led to the death of two mice—one each on days 7 and 10 None of the mice receiving 100 ng died
- mice receiving 0.1 ⁇ g, 0.5 ⁇ g, 1 ⁇ g, 2 ⁇ g, or 5 ⁇ g died and only one of four mice receiving 10 ⁇ g died on day 3 post-injection.
- the LD 50 was estimated at >10 micrograms.
- Plant RTA was significantly less toxic than either the wild-type ricin or mutant heterodimers.
- mice treated with RTA and the triple-site mutant ricin were calculated to be significantly different (p ⁇ 0.001) than the survival rates of wild-type, single-site mutant or double-site mutant ricins over a range of doses.
- mice treated with intraperitoneal ricin or RTA showed apoptosis in the thymus and spleen. Peritoneal inflammation was observed in some animals presumably due to intraperitoneal injection of toxin. There was no evidence of apoptosis based on nuclear morphology in the other organs examined. The triple-site mutant heterodimer treated mouse showed no histopathologic abnormalities.
- lymphoid apoptosis after ricin administration might suggest that those cells have a lower apoptotic threshold. Deletion of lymphocytes is an important ongoing process for tolerance induction and immune regulation in the lymphoid system (50).
- the toxicity of RTA alone may be due to the presence of small amounts
- EXAMPLE ⁇ i Construction of a ricin fusion protein immunotoxin containing a modification in a lectin binding site in each of the l ⁇ , l ⁇ and
- Transfer vector with mutant RTB was then purified by cesium chloride density centrifugation, restricted with BarnHI, bound and eluted from silica matrix (Promega, Madison, WI), digested with calf intestinal phosphatase (Boehringer- Mannheim, Indianapolis, IN), heat inactivated and repurified on silica matrix.
- the BamHI fragment encoding IL2 prepared by polymerase chain reaction of pDW27 plasmid DNA as previously described (54) was isolated from pUCl 19-
- LL2 by digestion of cesium chloride density gradient purified plasmid with BamHI, agarose electrophoresis and binding and elution from silica matrix.
- the 406 bp fragment was subcloned into pAcGP67A-ADP- RTB[W37S/Y248H/Y78H].
- the expression vector was maintained in INVaF E. coli using 100 ug/ml ampicillin. Plasmid isolated by alkaline lysis followed by cesium chloride density gradient centrifugation was double-stranded dideoxy sequenced by the Sanger method (33) using the Sequenase kit (USB, Cleveland, OH).
- fusion toxin Sf9 Spodoptera frugiperda ovarian cells (2 x 10 6 ), maintained in TMNFH medium supplemented with 10% fetal calf serum and 50 ug/ml gentamicin sulfate, were co-transfected with pAcGP67A-ADP- LL2-ADP-RTB[W37S/Y248H/Y78H] DNA (4 ⁇ g) and 0 5 ⁇ g of BaculoGold AcNPV DNA (PharMingen) following the recommendations of the supplier
- Recombinant virus in the supernatant was then amplified by infection Sf9 cells at an moi of 0 1, followed by collection of day 7 supernatants Recombinant baculovirus was then used to infect 2 x 10 8 Sf9 cells at an moi of 5-10 in 150 ml EXCELL400 medium (JRH Scientific, Lexena, KS) with 25 mM lactose in spinner flasks Media supernatants containing ADP-IL2-ADP-
- Protein purification Media supernatants were adjusted to 0 01% sodium azide and maintained through all purification steps at 4 n C The supernatants were concentrated 15-fold by vacuum dialysis, centrifuged at 3,000x g for 10 minutes to remove precipitate, dialyzed against 50 mM NaCl, 25 mM Tris (pH 8), 1 mM EDTA 0 01% sodium azide and 25 mM lactose (NTEAL), ultracentrif ⁇ iged at 100,000g for one hour and bound and eluted from a P2 monoclonal antibody-acrylamide matrix as previously described (53)
- P2 is an anti-RTB monoclonal antibody
- the affinity matrix was prepared using Ultralink azlactone functionality bis-acrylamide following the recommendations of the manufacturer (Pierce, Rockford, LL) Recombinant protein was absorbed to the column in NTEAL, washed with 0 5 M NaCl, 25 mM Tris pH 9, 1 mM EDTA 0.1% Tween 20, 0.02% sodium azide, 25 mM lactose and eluted with 0.1 M triethylamine hydrochloride (pH 11). The eluant was neutralized with 1/10 volume 1 M sodium phosphate pH 4.25 and stored at -20°C until assayed. Three preparations were made.
- Costar EIA microtiter wells were coated with 100 ⁇ l of 5 ug/ml of monoclonal antibody P2, P8, or P10 reactive with RTB or monoclonal antibody to LL2, washed with PBS plus 0.1% Tween 20, blocked with 3% BSA rewashed and incubated with samples of ADP-LL2-ADP-RTB[W37S/Y248H/Y78H], human IL2 or plant RTB, rewashed, reacted with 1 :400 rabbit antibody to ricin or 1 :500 rabbit antibody to LL2, washed again, incubated with 1 :5000 alkaline phosphatase conjugated goat anti-(rabbit IgG), rewashed, developed with 1 mg/ml p- nitrophenylphosphate in diethanolamine buffer (pH 9.6) and read on a BioRad 450 Microplate reader at 405 nm. Aliquots of ADP-LL2-ADP-
- RTB[W37S/Y248H Y78H], bacterial IL2, recombinant RTB, plant RTB, and prestained low molecular weight protein standards were run on a reducing 15% SDS-PAGE, transferred to nitrocellulose, blocked with 10% Carnation's nonfat dry milk/0.1% bovine serum albumin (BSA)/0.1% Tween 20, washed with PBS plus 0 05% Tween 20, reacted with either 1 400 rabbit antibody to ricin or 1 100 mouse monoclonal antibody to IL2 (5 ug/ml), rewashed, incubated with alkaline phosphatase conjugated goat anti-(rabbit IgG) or anti-(mouse IgG), washed again and developed with the Vectastain alkaline phosphatase kit
- HUT 102 human T leukemia cells bearing the high affinity LL2 receptor, YT2C2 human leukemia cells bearing the intermediate affinity LL2 receptor, MT-1 human leukemia cells bearing the low affinity IL2 receptor and CEM human leukemia cells and KB human epidermoid carcinoma cells lacking the IL2 receptor were washed with
- the cells were then washed with PBS and reacted with goat anti-(mouse Ig) conjugated to rhodamine (Jackson ImmunoResearch, West Grove, PA) at 25 ug/ml for 30 minutes at 4°C.
- the cells were washed again in PBS and fixed in
- Cytotoxicity assay Measurement of protein synthesis inhibition by ricin and ADP-IL2-ADP-RTB[W37S/Y248H/Y78H]-RTA in cultured cells was done as previously described using HUT 102, CEM, YT2C2, MT-1, and KB cells (15). ADP-LL2-ADP-wild-type RTB-RTA and ADP-LL2-ADP- RTB[W37S/Y248H] prepared as described previously were also tested. 18 ' 22 All assays were performed in triplicate. Twelve different concentrations of toxins were used. The LD 50 was the concentration of protein which inhibited protein synthesis by 50% compared with control wells without toxin. There was no purification step after heterodimer reassociation.
- the free RTA concentration at the highest concentration of heterodimer in the assay was 10 "7 M.
- the LD 50 for free RTA was 2 - 3 x 10 "6 M. 28
- the free RTA concentration (2 x 10 "9 M - 2 x 10 "13 M) should not produce cytotoxicity.
- HUT 102 cells ( 1.5 x 10 4 ) were placed in sterile Eppendorf tubes at 4°C in 100 ⁇ l leucine-poor RPMI
- 3 H-leucine was added as above and cells were harvested four hours later with a Skatron cell harvester and 3 H-leucine incorporation was measured in a liquid scintillation counter. Blocking of selective cytotoxicity was estimated by comparing the ID 50 of toxins in the presence or absence of
- P2 antibody ELISA showed the concentration of anti-RTB immunoreactive protein was 102 ⁇ g, 49 ⁇ g, and 75 ⁇ g, respectively. Thus, purity was between 21 and 85 % based on absorbance, BioRad protein assay and densitometry of Coomassie-stained gels. Immunologic cross-reactivity of ADP-IL2-ADP- RTB/W37S/Y248H/Y78H].
- the ricin Kd was 4 x 10 "9 M and the LL2 fusion toxin Kd was 1 2 x 10 "7 M Specificity for high affinity LL2 receptor was demonstrated on a live cell immunofluorescence assay The LL2 fusion toxin bound to HUT 102, YT2C2, MT-1 but did not bind CEM or KB cells. Binding to HUT 102 cells was inhibited by LL2 but not asialofetuin.
- the LL2- triple-site RTB mutant-RTA had improved specificity with IC 50 values of 5 x 10 "12 M on HUT 102 cells, 1 x 10 "9 M on CEM cells and 6 x 10 "10 M on KB cells.
- the in vitro therapeutic window (the ratio of the IC 50 of receptor negative cells to the IC 50 of receptor positive cells) was 1 for IL2-wild-type RTB-RTA 50 for LL2-RTB[W37S/Y248H]-RTA and 120 - 200 for IL2-
- LL2 receptor-mediated cell toxicity was tested by blocking experiments with excess LL2 or lactose. Excess IL2 reduced LL2-triple-site RTB mutant- RTA toxicity towards HUT 102 cells by 1 , 000-fold (IC 50 was 1 x 10 "8 M with
- IL2R targeted ricin fusion protein for preclinical and clinical development requires adequate yields, simple purification, adequate stability at room temperature and 37°C, and selective toxicity to IL2R bearing lymphocytes.
- the IL2 triple-site mutant RTB fusion protein was obtained at 50% purity in good yields of 0.75 mg/liter culture. This compares with 1 mg/liter for LL2 wild-type RTB and 0.34 mg/liter LL2 double-site mutant RTB fusion protein.
- the IL2 triple-site mutant RTB molecules reacted with antibodies to LL2 and RTB both by ELISA and Western blots. These results are evidence of proper folding of both the IL2 and RTB domains.
- the protein was secreted into the insect cell medium, and purification was accomplished by a one-step immunoaffinity absorption. This contrasts with the requirement for denaturation and refolding for bacterial toxin fusion proteins and chemical derivatization and conjugation for immunotoxins.
- the toxophore domain of ricin (RTA) was added by simply mixing with the LL2 triple-site mutant RTB at 10 "6 M. Extensive ionic and hydrophobic bonds in the RTA-RTB interface promote reassociation and disulfide bond formation (13). The observation of 80% reassociation compares favorably with the 60% reassociation for IL2 wild-type RTB-RTA (54), 55% reassociation for IL2 double-site mutant RTB-RTA and 50% reassociation for plant RTB-RTA under identical conditions (53). The heterodimers were stable at high dilution (10-12 M) suggesting formation of the disulfide bond between RTA Cys-259 and RTB Cys-4 (55).
- Binding specificity of the lectin-deficient heterodimer was demonstrated in both ELISA and cell immunofluorescence formats.
- the fusion toxin displayed 0.3% binding to immobilized asialofetuin.
- the Kd was 1.2 x 10 "7 M versus 4 x 10 "9 M for plant ricin. This weak binding compares to 1% binding
- Subdomain l ⁇ mutation W37S reduced sugar binding avidity 4-fold, while other 1 ⁇ subdomain mutations (K40M and K40M/N46G) yielded proteins with 7-8-fold reductions in asialofetuin avidity (40).
- the W37S mutation was used in the LL2-triple-site mutant because of its much better yields.
- the lack of detectable binding to other human cell lines which still have cell surface galactosides may be due to the insensitivity of the immunofluorescence assay, a two and one-half log reduction in sugar binding may be beneath the immunofluorescence detection limit. Binding to cell lines was blocked by IL2 but not asialofetuin.
- LL2-lectin-deficient ricin fusion protein The most important property of an LL2-lectin-deficient ricin fusion protein is its selective cytotoxicity to IL2R expressing cells.
- the triply cross-linked molecules lacked sugar-binding and were unable to intoxicate antigen-bearing cells.
- the lectin-deficient heterodimer was nontoxic to mouse macrophages in the presence of lactose, even though binding and internalization was mediated by binding to mannose receptors.
- the lectin-deficient ricin conjugate again had reduced cell cytotoxicity, although cell binding and entry was mediated by non- galactoside mechanisms.
- the IL2 -lectin-deficient fusion toxin was selectively cytotoxic to hematopoietic neoplastic cell lines with the heterotrimeric high affinity IL2R.
- the molecules were less toxic to cells with intermediate or low affinity LL2R, and nontoxic to cells without 1L2R. These results are similar to those seen with LL2-PE40 35 but distinct from those seen with DAB 389 LL2. The latter molecule fails to intoxicate low affinity LL2R ⁇ , ⁇ cells perhaps due to steric effects of the N-terminal toxin moiety. As a note of caution, fresh leukemic blasts often display lower levels of IL2R ⁇ and IL2R ⁇ than cell lines and may show lowered sensitivity to the ricin fusion molecule (23).
- the fusion toxin needs IL2R binding for cell intoxication.
- EXAMPLE TV Studies of mannose receptor mediated cell cytotoxicity of ricin fusion protein immunotoxins containing a modification in a lectin binding site in each of the l ⁇ , l ⁇ and 2 ⁇ subdomains.
- toxins Preparation of toxins.
- Recombinant baculovirus encoding the gp67A leader and the RTB mutant (W37S/Y78H/Y248H) was prepared as previously described and used to infect Sf9 insect cells at an moi of 5-10 in EXCELL400 medium (JRH Scientific, Lexena, KS) supplemented with 25 mM lactose.
- W37SiY78H/Y248H RTB protein was purified from day 6 post-infection cell supernatants by, sequentially, vacuum dialysis, dialysis into 50 mM NACL, 25 mM Tris HCL pH 8, 1 mM EDTA, 0. 1% sodium azide, and 25 mM lactose, binding and elution from a P2 monoclonal antibody anti-RTB azlactone functionality bis-acrylamide affinity matrix using 0. 1 M triethylamine HCL (pH 11) elution buffer and neutralization with 11 M NAPO, pH 4.
- Protein was quantitated by P2 antibody ELISA and reassociated overnight at room temperature with a three-fold molar excess of plant RTA (Inland Laboratories, Austin, TX). Heterodimer concentration was determined by a sandwich ELISA employing P2 monoclonal anti-RTB antibody capture and biotinylated ⁇ BR12 anti-RTA monoclonal antibody detection. Plant ricin was obtained from Inland Laboratories.
- Pansorbin fixed Staphyloccocus aureus Cowan strain was prewashed with 3% BSA in PBS and then incubated with cell extracts to remove non-specifically-binding labeled materials.
- the metabolically-labeled solutions were then reacted with Pansorbin pretreated with rabbit anti-ricin antibody.
- the Pansorbin was then pelleted and washed with RJPAL twice and RIPAL plus 500 mM NaCl once.
- the pellets were then boiled in reducing sample buffer and the samples run along with prestained low molecular standards on a 15% SDS polyacrylamide gel
- the gel was fixed briefly with 10% acetic acid, soaked for one hour in Enlightening, dried, exposed to x-ray film with enhancing screens for 1-3 days at -70 °C and developed
- Macrophage cells Mouse peritoneal macrophages were prepared using 12 week old female Balb/c mice house in an IACUC approved facility Briefly, mice were injected intraperitoneally with 1 5 ml Brewer's thioglycoUate medium (Becton Dickinson Microbiology Systems) After five days, mice were sacrificed The mice were then injected into their thigh fatty deposits with 10 ml of cold HEPES buffered Hank's balanced salt solution with 10 U/ml heparin with 11% BSA After injection, the mice were gently shaken and the injected medium withdrawn The cell suspension was centrifuged at l,000x g for 10 minutes and the cell pellet resuspended in 10 ml of RPMI 1640 with 10% fetal calf serum and 0 01 M HEPES Cells were then diluted to 2 x 10 6 / ml and seeded to 96 well plates at 100 ⁇ l/well After incubation for two hours at 37°C/5% CO
- the J774E mouse macrophage cell line (56) was cultured in ⁇ -MEM with 10% fetal calf serum, 60 ⁇ thioguanine and transferred from flasks to wells and dishes by exposure to trypsin EDTA (Gibco) Cells were plated at 2 x 10 cells/well in 96 well plates and 2 x 10 cells/35 mm dish, and incubated a further 24 hours at 37°C/5% C0 2 prior to assay MMR61 rat fibroblasts transfected with mouse mannose receptor cDNA were grown in Dulbecco's MEM containing 10% fetal calf serum and 400 ug/ml G418 (57). Cells were split and transferred to wells and dishes again by trypsinization and incubated a further 24 hours prior to assay.
- KB human epidermoid carcinoma cells obtained from the American Type Culture Collection (Rockville, MD) were grown in Dulbecco's MEM with 10% fetal calf serum (58). Cells were removed from flasks by trypsin treatment and seeded for experiments identically to the other cell types.
- the cells were again incubated four hours at 37°C/5% C0 2 and then harvested with a Skatron cell harvester onto glass fiber filter mats. Filters were dried and counted in Econofluor liquid scintillation fluid in a LKB liquid scintillation counter. The IC 50 was determined for each toxin/cell type/medium condition as the toxin concentration that reduced protein synthesis to 50% of control. Each assay was performed in quadruplicate. The mannose receptor directed toxicity was quantitated by the ratio of the toxin IC 50 in the presence of lactose plus mannan to the IC 50 in the presence of lactose alone. KB cells were assayed identically.
- J774E and MMR61 were assayed identically except leucine-free DMEM medium was used instead of RPMI 1640.
- Immunofluorescence assay Mouse peritoneal macrophages, J774E, MMR61 and KB cells were attached for 24 hours at 37° C in 5%C0 2 to petri dishes, fixed in 3.7% formaldehyde in PBS (15 minutes), washed with 2 mg/ml BSA in PBS and 0.1% saponin and incubated in PBS plus BSA with rabbit anti-mouse mannose receptor antibody (56) for 30 minutes at 4°C.
- the cells were rewashed with PBS and reacted with goat anti-(rabbit Ig) conjugated to rhodamine (Jackson ImmunoResearch, West Grove, PA) at 25 ug/ml with 0.1% saponin for 30 minutes at 23 °C. After a final wash with PBS, the cells were fixed in 3.7% formaldehyde in PBS, mounted under a #1 coverslip in glycerol-PBS (90: 10) and examined using a Zeiss Axioplan epifluorescence microscope (63 x, NA. 1.4 planapochromat objective). Fluorescence images were recorded using Tri-X film and negatives were digitized and edited using Adobe Photoshop software and a PowerMac 8500/120 computer. Relative intensities for the brightness of mannose receptor reactions in the different cell types were, respectively, for K-B, for J774E cells, and (+) for MMR61 and mouse peritoneal macrophages [scale (-) - (++++)].
- ricin with lactose on mouse peritoneal macrophages, J774E cells and MMR61 rat fibroblasts transfected with mouse mannose receptor were 8 x 10 "11 M, 1 x 10 " 10 M, and 8 x 10 "10 M, respectively.
- the triple-site mutant RTB-RTA with lactose showed IC 50 's on mouse peritoneal macrophages, J774E cells and MM61 cells of 1 x 10 "9 M, 3 x 10 "10 M, and 7 x 10 "9 M, respectively.
- the receptor-positive cells were 4 - 36 fold more sensitive to ricin and 30-100 fold more sensitive to triple-site mutant RTB-RTA heterodimer than receptor- negative cells.
- Ricin binds cell surface galactosides in the absence of lactose and intoxicates cells with an IC 50 of 1 X 10 "12 , 11 x 10 '11 M and 2 x 10 "11 M on mouse peritoneal macrophages, J774E cells and MMR61 cells, respectively.
- ricin was 10-80 fold more potent in the absence of lactose.
- the lectin-deficient mutant ricin had minimal residual galactoside mediated cytotoxicity with IC 50 'S of 5 x l0 " M, 7 x 10 " M, and 3 x 10 " M on mouse peritoneal macrophages,
- KB cells showed no effects of mannan on lectin deficient ricin toxicity in the absence of lactose (IC 50 of 1 x 10 " M for both) or presence of lactose (IC 50 of 3 x l0 "8 M for both).
- Lectin-deficient ricin behaved like ricin with significant mannan inhibition of toxicity on these cell lines. Mannan increased the IC 50 to 5 x 10 "9 M, 2 x 10 "8 M, and 3 x 10 "8 M for mouse peritoneal macrophages, J774E cells and MMR61 cells, respectively. This yielded a 4 - 50 fold reduction in toxicity.
- the lower potency of mannose receptor-mediated toxicity for both lactose blocked ricin and lectin-deficient ricin may be due to reduced surface receptor content or lower avidity of cell surface binding for the D-mannose receptors and may not reflect altered intracellular processing.
- the number of galactosyl-terminated cell surface glycoprotein receptors for ricin has been reported to be about 10 7 /mammalian cell (59) versus 105 /cell for D-mannose receptors (56). Avidities for each were similar with Ka's of 10 9 M "1 (56,59).
- J774E cells The greater sensitivity of J774E cells relative to MMR61 or mouse peritoneal macrophages may be due to higher cell surface mannose receptor density on J774E cells or different intracellular metabolism.
- the immunofluorescence assay suggests higher receptor content is the cause for the
- RTB lectin function for mannose receptor has also been documented for the IL2 receptor using an insect-derived lectin-deficient LL2 ricin fusion molecule.
- RTB enhancement for other receptors may be due to inefficient internalization by the ligand-receptor complex or misrouting intracellularly away from a translocation competent compartment (61,62).
- IC 50 is the concentration of toxin reducing protein synthesis by 50% after 24 hour incubation.
- LL2- W.T. RICIN LL2-wild-type RTB-RTA;
- IL2-D.S. RIC ⁇ N LL2-RTB[W37S/Y248H]- RTA;
- IL2-T.S. RICIN IL2-RTB[W37S/Y248H/Y78H]-RTA.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Gastroenterology & Hepatology (AREA)
- Immunology (AREA)
- Zoology (AREA)
- Toxicology (AREA)
- Botany (AREA)
- Peptides Or Proteins (AREA)
Abstract
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU50035/97A AU5003597A (en) | 1996-10-28 | 1997-10-28 | Methods and compositions for ricin fusion protein immunotoxins to treat cancer and autoimmune disease |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US73914796A | 1996-10-28 | 1996-10-28 | |
US08/739,147 | 1996-10-28 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO1998018820A1 true WO1998018820A1 (fr) | 1998-05-07 |
Family
ID=24971037
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US1997/019577 WO1998018820A1 (fr) | 1996-10-28 | 1997-10-28 | Procedes et compositions relatifs a des immunotoxines de la proteine de fusion du ricin, pour le traitement du cancer et de maladies auto-immunes |
Country Status (2)
Country | Link |
---|---|
AU (1) | AU5003597A (fr) |
WO (1) | WO1998018820A1 (fr) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1998029540A2 (fr) * | 1997-01-02 | 1998-07-09 | B.R.A.I.N. Biotechnology Research And Information Network Gmbh | Proteines recombinantes de fusion a base de proteines du gui viscum album qui desactivent les ribosomes |
WO2000058456A2 (fr) * | 1999-03-30 | 2000-10-05 | Board Of Regents, The University Of Texas System | Compositions et methodes permettant de modifier les effets toxiques de composes proteiques |
WO2000066755A2 (fr) * | 1999-04-28 | 2000-11-09 | Plant Bioscience Limited | Fusions pesticides |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1989001037A1 (fr) * | 1987-07-24 | 1989-02-09 | Cetus Corporation | Production de toxines de ricine dans un systeme d'expression de cellules d'insectes-baculovirus |
WO1989004839A1 (fr) * | 1987-11-24 | 1989-06-01 | Genetics Institute, Inc. | Molecules de ricin et conjugues de toxines de ricin ameliores |
-
1997
- 1997-10-28 AU AU50035/97A patent/AU5003597A/en not_active Abandoned
- 1997-10-28 WO PCT/US1997/019577 patent/WO1998018820A1/fr active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1989001037A1 (fr) * | 1987-07-24 | 1989-02-09 | Cetus Corporation | Production de toxines de ricine dans un systeme d'expression de cellules d'insectes-baculovirus |
WO1989004839A1 (fr) * | 1987-11-24 | 1989-06-01 | Genetics Institute, Inc. | Molecules de ricin et conjugues de toxines de ricin ameliores |
Non-Patent Citations (8)
Title |
---|
BURBAGE C. ET AL.,: "Rcin fusion toxin targeted to the human granulocyte-macrophage colony stimulating factor receptor is selectively toxic to acute myeloid leukemia cells", LEUKEMIA RESEARCH, vol. 21, no. 7, - July 1997 (1997-07-01), pages 681 - 690, XP002059513 * |
FRANKEL A. E. ET AL.,: "IL2 fused to lectin-deficient ricin is toxic to human leukemia cells expressing the IL2 receptor", LEUKEMIA, vol. 11, - January 1997 (1997-01-01), pages 22 - 30, XP002059512 * |
FRANKEL A. E. ET AL.,: "Ricin toxin contains at least three galactose-binding sites located in B chain subdomains 1alpha,1beta, and 2gamma", BIOCHEMISTRY, vol. 35, - 26 November 1996 (1996-11-26), pages 14749 - 14756, XP002059514 * |
FRANKEL A. ET AL.,: "Double-site ricin B chain mutants retain galactose binding", PROTEIN ENGINEERING, vol. 9, no. 4, - April 1996 (1996-04-01), pages 371 - 379, XP002059508 * |
FRANKEL A. ET AL.,: "IL-2 ricin fusion toxin is selectively cytotoxic in vitro to IL2 receptor-bearing tumor cells", BIOCONJ. CHEM., vol. 6, no. 6, - 1995, pages 666 - 672, XP002059509 * |
RUTENBER E. & ROBERTUS J. D.: "Structure of Ricin B-chain at 2.5A resolution", PROTEINS, vol. 10, - 1991, pages 260 - 269, XP002059515 * |
SPHYRIS N. ET AL.,: "Mutational analysis of the ricin lectin B-chains", J. BIOL. CHEM., vol. 270, no. 35, - 1 September 1995 (1995-09-01), pages 20292 - 20297, XP002059510 * |
WILLIAMS D. P. ET AL.,: "Diptheria toxin receptor binding domain substitution with interleukin-2: genetic construction and properties of a diphteria toxin-related interleukin-2 fusion protein", PROTEIN ENGINEERING, vol. 1, no. 6, - 1987, pages 493 - 498, XP002059511 * |
Cited By (10)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1998029540A2 (fr) * | 1997-01-02 | 1998-07-09 | B.R.A.I.N. Biotechnology Research And Information Network Gmbh | Proteines recombinantes de fusion a base de proteines du gui viscum album qui desactivent les ribosomes |
WO1998029540A3 (fr) * | 1997-01-02 | 1998-11-12 | B R A I N Biotechnology Resear | Proteines recombinantes de fusion a base de proteines du gui viscum album qui desactivent les ribosomes |
WO2000058456A2 (fr) * | 1999-03-30 | 2000-10-05 | Board Of Regents, The University Of Texas System | Compositions et methodes permettant de modifier les effets toxiques de composes proteiques |
WO2000058456A3 (fr) * | 1999-03-30 | 2001-02-15 | Univ Texas | Compositions et methodes permettant de modifier les effets toxiques de composes proteiques |
US6566500B1 (en) | 1999-03-30 | 2003-05-20 | Board Of Regents, The University Of Texas System | Compositions and methods for modifying toxic effects of proteinaceous compounds |
US7829668B2 (en) | 1999-03-30 | 2010-11-09 | Board Of Regents, The University Of Texas System | Compositions and methods for modifying toxic effects of proteinaceous compounds |
WO2000066755A2 (fr) * | 1999-04-28 | 2000-11-09 | Plant Bioscience Limited | Fusions pesticides |
WO2000066755A3 (fr) * | 1999-04-28 | 2001-01-18 | Plant Bioscience Ltd | Fusions pesticides |
AU777669B2 (en) * | 1999-04-28 | 2004-10-28 | Fraunhofer-Gesellschaft Zur Forderung Der Angewandten Forschung E.V. | Pesticidal fusions |
US7019197B1 (en) | 1999-04-28 | 2006-03-28 | Fraunhofer-Gesellschaft Zur Foerderung Der Angewandten Forschung E. V | Pesticidal fusions |
Also Published As
Publication number | Publication date |
---|---|
AU5003597A (en) | 1998-05-22 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2018372167B2 (en) | Partial agonists of interleukin-2 | |
FI120042B (fi) | Menetelmä kasvainkuoliotekijän reseptoria sisältävien fuusioproteiinien valmistamiseksi ja niitä koodaavia polynukleotideja ja vektoreita | |
KR20130091346A (ko) | 암 및 만성 감염 치료를 위한, 효능제 활성을 갖는 인터루킨 2로부터 유도된 폴리펩티드 | |
TW200539891A (en) | Methods of modulating cytokine activity; related reagents | |
JPH04218000A (ja) | 修飾ポリペプチド | |
JPH01502397A (ja) | M―csfの生産方法 | |
KR100439290B1 (ko) | 인터로이칸-18을인식하는신규의폴리펩티드 | |
JP2008525424A (ja) | 複合生成物 | |
WO2009117117A1 (fr) | Protéines hybrides et conjugués de récepteurs de lymphocytes t et procédés d'utilisation de ceux-ci | |
JPH08507201A (ja) | リンホトキシン‐β、リンホトキシン‐β複合体、それらの薬学的な調製物および治療への使用 | |
US20220213163A1 (en) | Il-2 chimeric proteins for immunosuppression | |
AU758576B2 (en) | Chemokines with amino-terminal modifications | |
US20080292628A1 (en) | Chimeric Protein | |
US7700318B2 (en) | Chimeric polypeptide and use thereof | |
TW202143996A (zh) | 使用可溶性cd24治療病毒性肺炎之方法 | |
Woo et al. | GMP production and characterization of the bivalent anti-human T cell immunotoxin, A-dmDT390-bisFv (UCHT1) for phase I/II clinical trials | |
WO1998018820A1 (fr) | Procedes et compositions relatifs a des immunotoxines de la proteine de fusion du ricin, pour le traitement du cancer et de maladies auto-immunes | |
JP2000515007A (ja) | T細胞選択的インターロイキン―4アゴニスト | |
AU2001283304B2 (en) | Method and composition for altering a T cell mediated pathology | |
Frankel et al. | IL2 fused to lectin-deficient ricin is toxic to human leukemia cells expressing the IL2 receptor | |
EP4386011A1 (fr) | Protéine de fusion d'interféron alpha d'anticorps ciblant gpc3 et son utilisation | |
AU2001283304A1 (en) | Method and composition for altering a T cell mediated pathology | |
KR20230096981A (ko) | 변형된 il-2 분자 및 이의 용도 | |
AU2005203503A1 (en) | Method and composition for altering a T cell mediated pathology |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A1 Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH HU IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A1 Designated state(s): GH KE LS MW SD SZ UG ZW AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
REG | Reference to national code |
Ref country code: DE Ref legal event code: 8642 |
|
NENP | Non-entry into the national phase |
Ref country code: JP Ref document number: 1998520712 Format of ref document f/p: F |
|
122 | Ep: pct application non-entry in european phase |